1
|
Riols F, Witting M, Haid M. Differential Mobility Spectrometry-Based Cardiolipin Analysis. Methods Mol Biol 2025; 2855:373-385. [PMID: 39354319 DOI: 10.1007/978-1-0716-4116-3_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Cardiolipins (CL) are special lipids in many respects. First of all, CL are composed of four fatty acids linked by two phosphatidic acids, which provide CL a unique molecular structure. Secondly, in eukaryotic cells they are specific to a single organelle, mitochondria, where they are also synthetized. CL are one of the most abundant lipid classes in mitochondria, mainly localized in the inner membrane. They are key determinants of mitochondrial health and homeostasis by modulating membrane integrity and fluidity, mitochondrial shapes, and metabolic pathways. Disturbances in mitochondrial CL composition can lead to tissue malfunction and diseases. It is therefore important to develop analytical tools to study the mitochondrial lipidome, and more particularly the CL. The method described here allows the quantification of cardiolipins at the sum composition level in isolated mitochondria or in liver tissue by flow injection analysis coupled to differential mobility spectrometry (FIA-DMS), also known as DMS-based shotgun lipidomics.
Collapse
Affiliation(s)
- Fabien Riols
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Neuherberg, Germany
| | - Michael Witting
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Mark Haid
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
2
|
Qi L, Liu S, Fang Q, Qian C, Peng C, Liu Y, Yang P, Wu P, Shan L, Cui Q, Hua Q, Yang S, Ye C, Yang W, Li P, Xu X. Ginsenoside Rg3 Restores Mitochondrial Cardiolipin Homeostasis via GRB2 to Prevent Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403058. [PMID: 39159293 PMCID: PMC11497058 DOI: 10.1002/advs.202403058] [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: 03/23/2024] [Revised: 06/20/2024] [Indexed: 08/21/2024]
Abstract
Regulating cardiolipin to maintain mitochondrial homeostasis is a promising strategy for addressing Parkinson's disease (PD). Through a comprehensive screening and validation process involving multiple models, ginsenoside Rg3 (Rg3) as a compound capable of enhancing cardiolipin levels is identified. This augmentation in cardiolipin levels fosters mitochondrial homeostasis by bolstering mitochondrial unfolded protein response, promoting mitophagy, and enhancing mitochondrial oxidative phosphorylation. Consequently, this cascade enhances the survival of tyrosine hydroxylase positive (TH+) dopaminergic neurons, leading to an amelioration in motor performance within PD mouse models. Using limited proteolysis-small-molecule mapping combined with molecular docking analysis, it has confirmed Growth Factor Receptor-Bound Protein 2 (GRB2) as a molecular target for Rg3. Furthermore, these investigations reveal that Rg3 facilitates the interaction between GRB2 and TRKA (Neurotrophic Tyrosine Kinase, Receptor, Type 1), thus promotes EVI1 (Ecotropic Virus Integration Site 1 Protein Homolog) phosphorylation by ERK, subsequently increases CRLS1 (Cardiolipin Synthase 1) gene expression and boosts cardiolipin synthesis. The absence of GRB2 or CRLS1 significantly attenuates the beneficial effects of Rg3 on PD symptoms. Finally, Tenofovir Disoproxil Fumarate (TDF) that also promotes the binding between GRB2 and TRKA is further identified. The identified compounds, Rg3 and TDF, exhibit promising potential for the prevention of PD by bolstering cardiolipin expression and reinstating mitochondrial homeostasis.
Collapse
Affiliation(s)
- Li‐Feng‐Rong Qi
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Shuai Liu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
- Department of PharmacyThe Fourth Affiliated HospitalCenter for Innovative Traditional Chinese Medicine Target and New Drug ResearchInternational Institutes of MedicineZhejiang University School of MedicineYiwuZhejiang322000China
| | - Qiuyuan Fang
- Department of Biophysics and Department of Neurosurgery of the First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Cheng Qian
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Chao Peng
- National Facility for Protein Science in ShanghaiZhangjiang LabShanghai Advanced Research InstituteChinese Academy of ScienceShanghai201210China
- Shanghai Science Research CenterChinese Academy of SciencesShanghai201204China
| | - Yuci Liu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Peng Yang
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Ping Wu
- National Facility for Protein Science in ShanghaiZhangjiang LabShanghai Advanced Research InstituteChinese Academy of ScienceShanghai201210China
- Shanghai Science Research CenterChinese Academy of SciencesShanghai201204China
| | - Ling Shan
- Dept. Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesMeibergdreef 47Amsterdam1105BAthe Netherlands
| | - Qinghua Cui
- Department of Biomedical InformaticsSchool of Basic Medical SciencesKey Laboratory of Molecular Cardiovascular Sciences of the Ministry of EducationCenter for Non‐Coding RNA MedicinePeking University Health Science Center BeijingBeijing100191China
| | - Qian Hua
- School of Life SciencesBeijing University of Chinese MedicineBeijing100029China
| | - Sen Yang
- Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Cunqi Ye
- Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Wei Yang
- Department of PharmacyThe Fourth Affiliated HospitalCenter for Innovative Traditional Chinese Medicine Target and New Drug ResearchInternational Institutes of MedicineZhejiang University School of MedicineYiwuZhejiang322000China
| | - Ping Li
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Xiaojun Xu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
- Department of PharmacyThe Fourth Affiliated HospitalCenter for Innovative Traditional Chinese Medicine Target and New Drug ResearchInternational Institutes of MedicineZhejiang University School of MedicineYiwuZhejiang322000China
| |
Collapse
|
3
|
Vieira Neto E, Wang M, Szuminsky AJ, Ferraro L, Koppes E, Wang Y, Van’t Land C, Mohsen AW, Zanatta G, El-Gharbawy AH, Anthonymuthu TS, Tyurina YY, Tyurin VA, Kagan V, Bayır H, Vockley J. Mitochondrial bioenergetics and cardiolipin remodeling abnormalities in mitochondrial trifunctional protein deficiency. JCI Insight 2024; 9:e176887. [PMID: 39088276 PMCID: PMC11385086 DOI: 10.1172/jci.insight.176887] [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: 10/30/2023] [Accepted: 07/25/2024] [Indexed: 08/03/2024] Open
Abstract
Mitochondrial trifunctional protein (TFP) deficiency is an inherited metabolic disorder leading to a block in long-chain fatty acid β-oxidation. Mutations in HADHA and HADHB, which encode the TFP α and β subunits, respectively, usually result in combined TFP deficiency. A single common mutation, HADHA c.1528G>C (p.E510Q), leads to isolated 3-hydroxyacyl-CoA dehydrogenase deficiency. TFP also catalyzes a step in the remodeling of cardiolipin (CL), a phospholipid critical to mitochondrial membrane stability and function. We explored the effect of mutations in TFP subunits on CL and other phospholipid content and composition and the consequences of these changes on mitochondrial bioenergetics in patient-derived fibroblasts. Abnormalities in these parameters varied extensively among different fibroblasts, and some cells were able to maintain basal oxygen consumption rates similar to controls. Although CL reduction was universally identified, a simultaneous increase in monolysocardiolipins was discrepant among cells. A similar profile was seen in liver mitochondria isolates from a TFP-deficient mouse model. Response to new potential drugs targeting CL metabolism might be dependent on patient genotype.
Collapse
Affiliation(s)
- Eduardo Vieira Neto
- Genetic and Genomic Medicine Division, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh
- Children’s Neuroscience Institute, Department of Pediatrics, School of Medicine, and
| | - Meicheng Wang
- Genetic and Genomic Medicine Division, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh
| | - Austin J. Szuminsky
- Department of Biological Sciences, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lethicia Ferraro
- Genetic and Genomic Medicine Division, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh
- School of Medicine and
| | - Erik Koppes
- Genetic and Genomic Medicine Division, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh
| | - Yudong Wang
- Genetic and Genomic Medicine Division, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh
| | - Clinton Van’t Land
- Genetic and Genomic Medicine Division, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh
| | - Al-Walid Mohsen
- Genetic and Genomic Medicine Division, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh
| | - Geancarlo Zanatta
- Department of Biophysics, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Areeg H. El-Gharbawy
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Yulia Y. Tyurina
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health
| | - Vladimir A. Tyurin
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health
| | - Valerian Kagan
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health
- Department of Pharmacology and Chemical Biology, School of Medicine; Department of Chemistry, Kenneth P. Dietrich School of Arts and Sciences; and Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hülya Bayır
- Division of Critical Care and Hospital Medicine, Department of Pediatrics, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Jerry Vockley
- Genetic and Genomic Medicine Division, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh
- Department of Human Genetics, School of Public Health, Center for Rare Disease Therapy, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Zhao P, Shi W, Ye Y, Xu K, Hu J, Chao H, Tao Z, Xu L, Gu W, Zhang L, Wang T, Wang X, Ji J. Atox1 protects hippocampal neurons after traumatic brain injury via DJ-1 mediated anti-oxidative stress and mitophagy. Redox Biol 2024; 72:103156. [PMID: 38640584 PMCID: PMC11047792 DOI: 10.1016/j.redox.2024.103156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024] Open
Abstract
Regulation of the oxidative stress response is crucial for the management and prognosis of traumatic brain injury (TBI). The copper chaperone Antioxidant 1 (Atox1) plays a crucial role in regulating intracellular copper ion balance and impacting the antioxidant capacity of mitochondria, as well as the oxidative stress state of cells. However, it remains unknown whether Atox1 is involved in modulating oxidative stress following TBI. Here, we investigated the regulatory role of Atox1 in oxidative stress on neurons both in vivo and in vitro, and elucidated the underlying mechanism through culturing hippocampal HT-22 cells with Atox1 mutation. The expression of Atox1 was significantly diminished following TBI, while mice with overexpressed Atox1 exhibited a more preserved hippocampal structure and reduced levels of oxidative stress post-TBI. Furthermore, the mice displayed notable impairments in learning and memory functions after TBI, which were ameliorated by the overexpression of Atox1. In the stretch injury model of HT-22 cells, overexpression of Atox1 mitigated oxidative stress by preserving the normal morphology and network connectivity of mitochondria, as well as facilitating the elimination of damaged mitochondria. Mechanistically, co-immunoprecipitation and mass spectrometry revealed the binding of Atox1 to DJ-1. Knockdown of DJ-1 in HT-22 cells significantly impaired the antioxidant capacity of Atox1. Mutations in the copper-binding motif or sequestration of free copper led to a substantial decrease in the interaction between Atox1 and DJ-1, with overexpression of DJ-1 failing to restore the antioxidant capacity of Atox1 mutants. The findings suggest that DJ-1 mediates the ability of Atox1 to withstand oxidative stress. And targeting Atox1 could be a potential therapeutic approach for addressing post-traumatic neurological dysfunction.
Collapse
Affiliation(s)
- Pengzhan Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenqian Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yangfan Ye
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ke Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingming Hu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Honglu Chao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - ZeQiang Tao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Gu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liuchao Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tian Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyue Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Ji
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Gusu School, Nanjing Medical University, Suzhou, China; Department of Neurosurgery, The Affiliated Kizilsu Kirghiz Autonomous Prefecture People's Hospital of Nanjing Medical University, Artux, Xinjiang, China.
| |
Collapse
|
5
|
Li Y, Deng H, Zhang H, Yang L, Wang S, Wang H, Zhu J, Li X, Chen X, Lin Y, Li R, Wang G, Li K. Transforming growth factor-β1 protects mechanically injured cortical murine neurons by reducing trauma-induced autophagy and apoptosis. Front Cell Neurosci 2024; 18:1381279. [PMID: 38863498 PMCID: PMC11165077 DOI: 10.3389/fncel.2024.1381279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
Transforming growth factor β1 (TGF-β1) has a neuroprotective function in traumatic brain injury (TBI) through its anti-inflammatory and immunomodulatory properties. However, the precise mechanisms underlying the neuroprotective actions of TGF-β1 on the cortex require further investigation. In this study, we were aimed to investigate the regulatory function of TGF-β1 on neuronal autophagy and apoptosis using an in vitro primary cortical neuron trauma-injury model. LDH activity was assayed to measure cell viability, and intracellular [Ca2+] was measured using Fluo-4-AM in an in vitro primary cortical neuron trauma-injury model. RNA-sequencing (RNAseq), immunofluorescent staining, transmission electron microscopy (TEM), western blot and CTSD activity detection were employed. We observed significant enrichment of DEGs related to autophagy, apoptosis, and the lysosome pathway in trauma-injured cortical neurons. TEM confirmed the presence of autophagosomes as well as autophagolysosomes. Western blot revealed upregulation of autophagy-related protein light chain 3 (LC3-II/LC3-I), sequestosome 1 (SQSTM1/p62), along with apoptosis-related protein cleaved-caspase 3 in trauma-injured primary cortical neurons. Furthermore, trauma-injured cortical neurons showed an upregulation of lysosomal marker protein (LAMP1) and lysosomal enzyme mature cathepsin D (mCTSD), but a decrease in the activity of CTSD enzyme. These results indicated that apoptosis was up-regulated in trauma- injured cortical neurons at 24 h, accompanied by lysosomal dysfunction and impaired autophagic flux. Notably, TGF-β1 significantly reversed these changes. Our results suggested that TGF-β1 exerted neuroprotective effects on trauma- injured cortical neurons by reducing lysosomal dysfunction, decreasing the accumulation of autophagosomes and autophagolysosomes, and enhancing autophagic flux.
Collapse
Affiliation(s)
- Yanlei Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Huixiong Deng
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Hengyao Zhang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Lin Yang
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong, China
| | - Shenmiao Wang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Haoyang Wang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jiacheng Zhu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaoning Li
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxuan Chen
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yinhong Lin
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Rui Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Gefei Wang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Kangsheng Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
6
|
Torp MK, Stensløkken KO, Vaage J. When Our Best Friend Becomes Our Worst Enemy: The Mitochondrion in Trauma, Surgery, and Critical Illness. J Intensive Care Med 2024:8850666241237715. [PMID: 38505947 DOI: 10.1177/08850666241237715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Common for major surgery, multitrauma, sepsis, and critical illness, is a whole-body inflammation. Tissue injury is able to trigger a generalized inflammatory reaction. Cell death causes release of endogenous structures termed damage associated molecular patterns (DAMPs) that initiate a sterile inflammation. Mitochondria are evolutionary endosymbionts originating from bacteria, containing molecular patterns similar to bacteria. These molecular patterns are termed mitochondrial DAMPs (mDAMPs). Mitochondrial debris released into the extracellular space or into the circulation is immunogenic and damaging secondary to activation of the innate immune system. In the circulation, released mDAMPS are either free or exist in extracellular vesicles, being able to act on every organ and cell in the body. However, the role of mDAMPs in trauma and critical care is not fully clarified. There is a complete lack of knowledge how they may be counteracted in patients. Among mDAMPs are mitochondrial DNA, cardiolipin, N-formyl peptides, cytochrome C, adenosine triphosphate, reactive oxygen species, succinate, and mitochondrial transcription factor A. In this overview, we present the different mDAMPs, their function, release, targets, and inflammatory potential. In light of present knowledge, the role of mDAMPs in the pathophysiology of major surgery and trauma as well as sepsis, and critical care is discussed.
Collapse
Affiliation(s)
- May-Kristin Torp
- Section of Physiology, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
- Department of Research, Østfold Hospital Trust, Grålum, Norway
| | - Kåre-Olav Stensløkken
- Section of Physiology, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
| | - Jarle Vaage
- Section of Physiology, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
- Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
7
|
Harris G, Stickland CA, Lim M, Goldberg Oppenheimer P. Raman Spectroscopy Spectral Fingerprints of Biomarkers of Traumatic Brain Injury. Cells 2023; 12:2589. [PMID: 37998324 PMCID: PMC10670390 DOI: 10.3390/cells12222589] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023] Open
Abstract
Traumatic brain injury (TBI) affects millions of people of all ages around the globe. TBI is notoriously hard to diagnose at the point of care, resulting in incorrect patient management, avoidable death and disability, long-term neurodegenerative complications, and increased costs. It is vital to develop timely, alternative diagnostics for TBI to assist triage and clinical decision-making, complementary to current techniques such as neuroimaging and cognitive assessment. These could deliver rapid, quantitative TBI detection, by obtaining information on biochemical changes from patient's biofluids. If available, this would reduce mis-triage, save healthcare providers costs (both over- and under-triage are expensive) and improve outcomes by guiding early management. Herein, we utilize Raman spectroscopy-based detection to profile a panel of 18 raw (human, animal, and synthetically derived) TBI-indicative biomarkers (N-acetyl-aspartic acid (NAA), Ganglioside, Glutathione (GSH), Neuron Specific Enolase (NSE), Glial Fibrillary Acidic Protein (GFAP), Ubiquitin C-terminal Hydrolase L1 (UCHL1), Cholesterol, D-Serine, Sphingomyelin, Sulfatides, Cardiolipin, Interleukin-6 (IL-6), S100B, Galactocerebroside, Beta-D-(+)-Glucose, Myo-Inositol, Interleukin-18 (IL-18), Neurofilament Light Chain (NFL)) and their aqueous solution. The subsequently derived unique spectral reference library, exploiting four excitation lasers of 514, 633, 785, and 830 nm, will aid the development of rapid, non-destructive, and label-free spectroscopy-based neuro-diagnostic technologies. These biomolecules, released during cellular damage, provide additional means of diagnosing TBI and assessing the severity of injury. The spectroscopic temporal profiles of the studied biofluid neuro-markers are classed according to their acute, sub-acute, and chronic temporal injury phases and we have further generated detailed peak assignment tables for each brain-specific biomolecule within each injury phase. The intensity ratios of significant peaks, yielding the combined unique spectroscopic barcode for each brain-injury marker, are compared to assess variance between lasers, with the smallest variance found for UCHL1 (σ2 = 0.000164) and the highest for sulfatide (σ2 = 0.158). Overall, this work paves the way for defining and setting the most appropriate diagnostic time window for detection following brain injury. Further rapid and specific detection of these biomarkers, from easily accessible biofluids, would not only enable the triage of TBI, predict outcomes, indicate the progress of recovery, and save healthcare providers costs, but also cement the potential of Raman-based spectroscopy as a powerful tool for neurodiagnostics.
Collapse
Affiliation(s)
- Georgia Harris
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Clarissa A. Stickland
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Matthias Lim
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Pola Goldberg Oppenheimer
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Institute of Healthcare Technologies, Mindelsohn Way, Birmingham B15 2TH, UK
| |
Collapse
|
8
|
Bautista JS, Falabella M, Flannery PJ, Hanna MG, Heales SJ, Pope SA, Pitceathly RD. Advances in methods to analyse cardiolipin and their clinical applications. Trends Analyt Chem 2022; 157:116808. [PMID: 36751553 PMCID: PMC7614147 DOI: 10.1016/j.trac.2022.116808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cardiolipin (CL) is a mitochondria-exclusive phospholipid, primarily localised within the inner mitochondrial membrane, that plays an essential role in mitochondrial architecture and function. Aberrant CL content, structure, and localisation have all been linked to impaired mitochondrial activity and are observed in the pathophysiology of cancer and neurological, cardiovascular, and metabolic disorders. The detection, quantification, and localisation of CL species is a valuable tool to investigate mitochondrial dysfunction and the pathophysiological mechanisms underpinning several human disorders. CL is measured using liquid chromatography, usually combined with mass spectrometry, mass spectrometry imaging, shotgun lipidomics, ion mobility spectrometry, fluorometry, and radiolabelling. This review summarises available methods to analyse CL, with a particular focus on modern mass spectrometry, and evaluates their advantages and limitations. We provide guidance aimed at selecting the most appropriate technique, or combination of techniques, when analysing CL in different model systems, and highlight the clinical contexts in which measuring CL is relevant.
Collapse
Affiliation(s)
- Javier S. Bautista
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Micol Falabella
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Padraig J. Flannery
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London, UK,Neurogenetics Unit, Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, London, UK
| | - Michael G. Hanna
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK,NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Simon J.R. Heales
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London, UK,NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK,Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Simon A.S. Pope
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London, UK,Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Robert D.S. Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK,NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK, Corresponding author. Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK. (R.D.S. Pitceathly)
| |
Collapse
|
9
|
Zhao P, Wei Y, Sun G, Xu L, Wang T, Tian Y, Chao H, Tu Y, Ji J. Fetuin-A alleviates neuroinflammation against traumatic brain injury-induced microglial necroptosis by regulating Nrf-2/HO-1 pathway. J Neuroinflammation 2022; 19:269. [PMID: 36333786 PMCID: PMC9636801 DOI: 10.1186/s12974-022-02633-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The microglia-mediated inflammatory response is a vital mechanism of secondary damage following traumatic brain injury (TBI), but the underlying mechanism of microglial activation is unclear. METHODS Controlled cortical impact (CCI) was induced in adult male C57BL/6J mice, and glutamate was used to construct a classical in vitro injury model in the primary microglia. Microglial activation was determined by western blot and immunostaining. The inflammatory factors were measured by enzyme-linked immunosorbent assay. The oxidative stress marker and mitochondrial reactive oxygen species (ROS) were measured by immunoblotting and MitoSox Red staining. Transmission electron microscopy was used to observe the typical morphology of necroptotic cells. RESULTS Our quantitative proteomics identified 2499 proteins; 157 were significantly differentially expressed in brain tissue between the 6 h after CCI (CCI6h) group and sham group, and 109 were significantly differentially expressed between the CCI24h and sham groups. Moreover, compared with the sham group, the terms "acute-phase response", "inflammation", and "protein binding" were significantly enriched in CCI groups. Fetuin-A, a liver-secreted acute-phase glycoprotein, was involved in these biological processes. Using an experimental TBI model, we found that the Fetuin-A level peaked at 6 h and then decreased gradually. Importantly, we showed that administration of Fetuin-A reduced the cortical lesion volume and edema area and inhibited the inflammatory response, which was associated with suppressing microglial necroptosis, thus decreasing microglial activation. Furthermore, administration of Fetuin-A attenuated mitochondrial oxidative stress in glutamate-treated microglial cells, which is a critical mechanism of necroptosis suppression. In addition, we demonstrated that Fetuin-A treatment promoted translocation of nuclear factor erythroid 2-related factor 2 (Nrf-2) from the cytoplasm to the nucleus in vivo; however, the Nrf-2 inhibitor ML385 and si-heme oxygenase-1 (si-HO-1) disrupted the regulation of oxidative stress by Fetuin-A and induced increased ROS levels and necroptosis in glutamate-treated microglial cells. Fetuin-A also protected neurons from adverse factors in vivo and in vitro. CONCLUSIONS Our results demonstrated that Fetuin-A activated Nrf-2/HO-1, suppressed oxidative stress and necroptosis levels, and thereby attenuates the abnormal inflammatory response following TBI. The findings suggest a potential therapeutic strategy for TBI treatment.
Collapse
Affiliation(s)
- Pengzhan Zhao
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China ,grid.412679.f0000 0004 1771 3402Department of Neurosurgery, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031 Anhui China
| | - Yutian Wei
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China
| | - Guangchi Sun
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China
| | - Lei Xu
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China
| | - Tian Wang
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China
| | - Yufei Tian
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China
| | - Honglu Chao
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China
| | - Yiming Tu
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China
| | - Jing Ji
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China ,grid.89957.3a0000 0000 9255 8984Gusu School, Nanjing Medical University, Suzhou, 215031 Jiangsu China
| |
Collapse
|
10
|
Jiang Z, Shen T, Huynh H, Fang X, Han Z, Ouyang K. Cardiolipin Regulates Mitochondrial Ultrastructure and Function in Mammalian Cells. Genes (Basel) 2022; 13:genes13101889. [PMID: 36292774 PMCID: PMC9601307 DOI: 10.3390/genes13101889] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/01/2022] Open
Abstract
Cardiolipin (CL) is a unique, tetra-acylated diphosphatidylglycerol lipid that mainly localizes in the inner mitochondria membrane (IMM) in mammalian cells and plays a central role in regulating mitochondrial architecture and functioning. A deficiency of CL biosynthesis and remodeling perturbs mitochondrial functioning and ultrastructure. Clinical and experimental studies on human patients and animal models have also provided compelling evidence that an abnormal CL content, acyl chain composition, localization, and level of oxidation may be directly linked to multiple diseases, including cardiomyopathy, neuronal dysfunction, immune cell defects, and metabolic disorders. The central role of CL in regulating the pathogenesis and progression of these diseases has attracted increasing attention in recent years. In this review, we focus on the advances in our understanding of the physiological roles of CL biosynthesis and remodeling from human patients and mouse models, and we provide an overview of the potential mechanism by which CL regulates the mitochondrial architecture and functioning.
Collapse
Affiliation(s)
- Zhitong Jiang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen 518055, China
| | - Tao Shen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen 518055, China
| | - Helen Huynh
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, San Diego, CA 92093, USA
| | - Xi Fang
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, San Diego, CA 92093, USA
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen 518055, China
- Correspondence: (Z.H.); (K.O.)
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen 518055, China
- Correspondence: (Z.H.); (K.O.)
| |
Collapse
|
11
|
Liu X, Zhang J, Li J, Song C, Shi Y. Pharmacological Inhibition of ALCAT1 Mitigates Amyotrophic Lateral Sclerosis by Attenuating SOD1 Protein Aggregation. Mol Metab 2022; 63:101536. [PMID: 35772643 PMCID: PMC9287437 DOI: 10.1016/j.molmet.2022.101536] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/11/2022] Open
Abstract
Objective Mutations in the copper-zinc superoxide dismutase (SOD1) gene cause familial amyotrophic lateral sclerosis (ALS), a progressive fatal neuromuscular disease characterized by motor neurons death and severe skeletal muscle degeneration. However, there is no effective treatment for this debilitating disease, since the underlying cause for the pathogenesis remains poorly understood. Here, we investigated a role of acyl-CoA:lysocardiolipin acyltransferase 1 (ALCAT1), an acyltransferase that promotes mitochondrial dysfunction in age-related diseases by catalyzing pathological remodeling of cardiolipin, in promoting the development of ALS in the SOD1G93A transgenic mice. Methods Using SOD1G93A transgenic mice with targeted deletion of the ALCAT1 gene and treated with Dafaglitapin (Dafa), a very potent and highly selective ALCAT1 inhibitor, we determined whether ablation or pharmaceutical inhibition of ALCAT1 by Dafa would mitigate ALS and the underlying pathogenesis by preventing pathological remodeling of cardiolipin, oxidative stress, and mitochondrial dysfunction by multiple approaches, including lifespan analysis, behavioral tests, morphological and functional analysis of skeletal muscle, electron microscopic and Seahorse analysis of mitochondrial morphology and respiration, western blot analysis of the SOD1G93A protein aggregation, and lipidomic analysis of cardiolipin content and acyl composition in mice spinal cord. Results ALCAT1 protein expression is potently upregulated in the skeletal muscle of the SOD1G93A mice. Consequently, ablation or pharmacological inhibition of ALCAT1 by Dafa attenuates motor neuron dysfunction, neuronal inflammation, and skeletal muscle atrophy in SOD1G93A mice by preventing SOD1G93A protein aggregation, mitochondrial dysfunction, and pathological CL remodeling, leading to moderate extension of lifespan in the SOD1G93A transgenic mice. Conclusions ALCAT1 promotes the development of ALS by linking SOD1G93A protein aggregation to mitochondrial dysfunction, implicating Dafa as a potential treatment for this debilitating disorder. ALCAT1 is potently upregulated in the skeletal muscle of SOD1G93A mice, a mouse model of amyotrophic lateral sclerosis. Upregulated ALCAT1 promotes SOD1G93A protein aggregation through oxidative stress and pathological cardiolipin remodeling. Inactivation of ALCAT1 attenuates neuronal mitochondrial dysfunction and extends the lifespan of SOD1G93A mice. Targeting ALCAT1 as a potential strategy for the treatment of amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Xueling Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China; Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jun Zhang
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Chengjie Song
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yuguang Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China; Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
12
|
Lupeol Treatment Attenuates Activation of Glial Cells and Oxidative-Stress-Mediated Neuropathology in Mouse Model of Traumatic Brain Injury. Int J Mol Sci 2022; 23:ijms23116086. [PMID: 35682768 PMCID: PMC9181489 DOI: 10.3390/ijms23116086] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 02/05/2023] Open
Abstract
Traumatic brain injury (TBI) signifies a major cause of death and disability. TBI causes central nervous system (CNS) damage under a variety of mechanisms, including protein aggregation, mitochondrial dysfunction, oxidative stress, and neuroinflammation. Astrocytes and microglia, cells of the CNS, are considered the key players in initiating an inflammatory response after injury. Several evidence suggests that activation of astrocytes/microglia and ROS/LPO have the potential to cause more harmful effects in the pathological processes following traumatic brain injury (TBI). Previous studies have established that lupeol provides neuroprotection through modulation of inflammation, oxidative stress, and apoptosis in Aβ and LPS model and neurodegenerative disease. However, the effects of lupeol on apoptosis caused by inflammation and oxidative stress in TBI have not yet been investigated. Therefore, we explored the role of Lupeol on antiapoptosis, anti-inflammatory, and antioxidative stress and its potential mechanism following TBI. In these experiments, adult male mice were randomly divided into four groups: control, TBI, TBI+ Lupeol, and Sham group. Western blotting, immunofluorescence staining, and ROS/LPO assays were performed to investigate the role of lupeol against neuroinflammation, oxidative stress, and apoptosis. Lupeol treatment reversed TBI-induced behavioral and memory disturbances. Lupeol attenuated TBI-induced generation of reactive oxygen species/lipid per oxidation (ROS/LPO) and improved the antioxidant protein level, such as nuclear factor erythroid 2-related factor 2 (Nrf2) and heme-oxygenase 1 (HO-1) in the mouse brain. Similarly, our results indicated that lupeol treatment inhibited glial cell activation, p-NF-κB, and downstream signaling molecules, such as TNF-α, COX-2, and IL-1β, in the mouse cortex and hippocampus. Moreover, lupeol treatment also inhibited mitochondrial apoptotic signaling molecules, such as caspase-3, Bax, cytochrome-C, and reversed deregulated Bcl2 in TBI-treated mice. Overall, our study demonstrated that lupeol inhibits the activation of astrocytes/microglia and ROS/LPO that lead to oxidative stress, neuroinflammation, and apoptosis followed by TBI.
Collapse
|
13
|
Mitophagy in Traumatic Brain Injury: A New Target for Therapeutic Intervention. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4906434. [PMID: 35126814 PMCID: PMC8813270 DOI: 10.1155/2022/4906434] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/31/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) contributes to death, and disability worldwide more than any other traumatic insult and damage to cellular components including mitochondria leads to the impairment of cellular functions and brain function. In neurons, mitophagy, autophagy-mediated degradation of damaged mitochondria, is a key process in cellular quality control including mitochondrial homeostasis and energy supply and plays a fundamental role in neuronal survival and health. Conversely, defective mitophagy leads to the accumulation of damaged mitochondria and cellular dysfunction, contributing to inflammation, oxidative stress, and neuronal cell death. Therefore, an extensive characterization of mitophagy-related protective mechanisms, taking into account the complex mechanisms by which each molecular player is connected to the others, may provide a rationale for the development of new therapeutic strategies in TBI patients. Here, we discuss the contribution of defective mitophagy in TBI, and the underlying molecular mechanisms of mitophagy in inflammation, oxidative stress, and neuronal cell death highlight novel therapeutics based on newly discovered mitophagy-inducing strategies.
Collapse
|
14
|
Antioxidant Role and Cardiolipin Remodeling by Redox-Activated Mitochondrial Ca 2+-Independent Phospholipase A 2γ in the Brain. Antioxidants (Basel) 2022; 11:antiox11020198. [PMID: 35204081 PMCID: PMC8868467 DOI: 10.3390/antiox11020198] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
Mitochondrial Ca2+-independent phospholipase A2γ (iPLA2γ/PNPLA8) was previously shown to be directly activated by H2O2 and release free fatty acids (FAs) for FA-dependent H+ transport mediated by the adenine nucleotide translocase (ANT) or uncoupling protein 2 (UCP2). The resulting mild mitochondrial uncoupling and consequent partial attenuation of mitochondrial superoxide production lead to an antioxidant effect. However, the antioxidant role of iPLA2γ in the brain is not completely understood. Here, using wild-type and iPLA2γ-KO mice, we demonstrate the ability of tert-butylhydroperoxide (TBHP) to activate iPLA2γ in isolated brain mitochondria, with consequent liberation of FAs and lysophospholipids. The liberated FA caused an increase in respiratory rate, which was fully inhibited by carboxyatractyloside (CATR), a specific inhibitor of ANT. Employing detailed lipidomic analysis, we also demonstrate a typical cleavage pattern for TBHP-activated iPLA2γ, reflecting cleavage of glycerophospholipids from both sn-1 and sn-2 positions releasing saturated FAs, monoenoic FAs, and predominant polyunsaturated FAs. The acute antioxidant role of iPLA2γ-released FAs is supported by monitoring both intramitochondrial superoxide and extramitochondrial H2O2 release. We also show that iPLA2γ-KO mice were more sensitive to stimulation by pro-inflammatory lipopolysaccharide, as reflected by the concomitant increase in protein carbonyls in the brain and pro-inflammatory IL-6 release in the serum. These data support the antioxidant and anti-inflammatory role of iPLA2γ in vivo. Our data also reveal a substantial decrease of several high molecular weight cardiolipin (CL) species and accumulation of low molecular weight CL species in brain mitochondria of iPLA2γ-KO mice. Collectively, our results support a key role of iPLA2γ in the remodeling of lower molecular weight immature cardiolipins with predominantly saturated acyl chains to high molecular weight mature cardiolipins with highly unsaturated PUFA acyl chains, typical for the brain.
Collapse
|
15
|
Human Cerebral Organoid Implantation Alleviated the Neurological Deficits of Traumatic Brain Injury in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6338722. [PMID: 34853630 PMCID: PMC8629662 DOI: 10.1155/2021/6338722] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) causes a high rate of mortality and disability, and its treatment is still limited. Loss of neurons in damaged area is hardly rescued by relative molecular therapies. Based on its disease characteristics, we transplanted human embryonic stem cell- (hESC-) derived cerebral organoids in the brain lesions of controlled cortical impact- (CCI-) modeled severe combined immunodeficient (SCID) mice. Grafted organoids survived and differentiated in CCI-induced lesion pools in mouse cortical tissue. Implanted cerebral organoids differentiated into various types of neuronal cells, extended long projections, and showed spontaneous action, as indicated by electromyographic activity in the grafts. Induced vascularization and reduced glial scar were also found after organoid implantation, suggesting grafting could improve local situation and promote neural repair. More importantly, the CCI mice's spatial learning and memory improved after organoid grafting. These findings suggest that cerebral organoid implanted in lesion sites differentiates into cortical neurons, forms long projections, and reverses deficits in spatial learning and memory, a potential therapeutic avenue for TBI.
Collapse
|
16
|
Villaseñor A, Godzien J, Barker-Tejeda TC, Gonzalez-Riano C, López-López Á, Dudzik D, Gradillas A, Barbas C. Analytical approaches for studying oxygenated lipids in the search of potential biomarkers by LC-MS. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
17
|
Bao Z, Liu Y, Chen B, Miao Z, Tu Y, Li C, Chao H, Ye Y, Xu X, Sun G, Zhao P, Liu N, Liu Y, Wang X, Lam SM, Kagan VE, Bayır H, Ji J. Prokineticin-2 prevents neuronal cell deaths in a model of traumatic brain injury. Nat Commun 2021; 12:4220. [PMID: 34244497 PMCID: PMC8270965 DOI: 10.1038/s41467-021-24469-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/18/2021] [Indexed: 01/20/2023] Open
Abstract
Prokineticin-2 (Prok2) is an important secreted protein likely involved in the pathogenesis of several acute and chronic neurological diseases through currently unidentified regulatory mechanisms. The initial mechanical injury of neurons by traumatic brain injury triggers multiple secondary responses including various cell death programs. One of these is ferroptosis, which is associated with dysregulation of iron and thiols and culminates in fatal lipid peroxidation. Here, we explore the regulatory role of Prok2 in neuronal ferroptosis in vitro and in vivo. We show that Prok2 prevents neuronal cell death by suppressing the biosynthesis of lipid peroxidation substrates, arachidonic acid-phospholipids, via accelerated F-box only protein 10 (Fbxo10)-driven ubiquitination, degradation of long-chain-fatty-acid-CoA ligase 4 (Acsl4), and inhibition of lipid peroxidation. Mice injected with adeno-associated virus-Prok2 before controlled cortical impact injury show reduced neuronal degeneration and improved motor and cognitive functions, which could be inhibited by Fbxo10 knockdown. Our study shows that Prok2 mediates neuronal cell deaths in traumatic brain injury via ferroptosis.
Collapse
Affiliation(s)
- Zhongyuan Bao
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yinlong Liu
- Department of Neurosurgery, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Binglin Chen
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zong Miao
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yiming Tu
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chong Li
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Honglu Chao
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yangfan Ye
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiupeng Xu
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guangchi Sun
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pengzhan Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ning Liu
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Liu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xiaoming Wang
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Sin Man Lam
- LipidALL Technologies Company Limited, Changzhou, China
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Heath, Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA.,Laboratory of Navigational Redox Lipidomics, IM Sechenov Moscow State Medical University, Moscow, Russian Federation
| | - Hülya Bayır
- Center for Free Radical and Antioxidant Heath, Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA.,Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Jing Ji
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
18
|
Olivar-Villanueva M, Ren M, Phoon CKL. Neurological & psychological aspects of Barth syndrome: Clinical manifestations and potential pathogenic mechanisms. Mitochondrion 2021; 61:188-195. [PMID: 34197965 DOI: 10.1016/j.mito.2021.06.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 06/10/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
Barth syndrome is a rare X-linked multisystem mitochondrial disease that is caused by variants in the tafazzin gene leading to deficient and abnormal cardiolipin. Previous research has focused on the cardiomyopathy and neutropenia in individuals with Barth syndrome, yet just as common are the least explored neurological aspects of Barth syndrome. This review focuses on the major neuropsychological and neurophysiological phenotypes that affect the quality of life of individuals with Barth syndrome, including difficulties in sensory perception and feeding, fatigue, and cognitive and psychological challenges. We propose selected pathogenetic mechanisms underlying these phenotypes and draw parallels to other relevant disorders. Finally, avenues for future research are also suggested.
Collapse
Affiliation(s)
- Melissa Olivar-Villanueva
- Departments of Pediatrics, New York University Grossman School of Medicine, New York, NY, United States
| | - Mindong Ren
- Departments of Anesthesiology, New York University Grossman School of Medicine, New York, NY, United States; Departments of Cell Biology, New York University Grossman School of Medicine, New York, NY, United States
| | - Colin K L Phoon
- Departments of Pediatrics, New York University Grossman School of Medicine, New York, NY, United States.
| |
Collapse
|
19
|
Chen D, Chu B, Yang X, Liu Z, Jin Y, Kon N, Rabadan R, Jiang X, Stockwell BR, Gu W. iPLA2β-mediated lipid detoxification controls p53-driven ferroptosis independent of GPX4. Nat Commun 2021; 12:3644. [PMID: 34131139 PMCID: PMC8206155 DOI: 10.1038/s41467-021-23902-6] [Citation(s) in RCA: 180] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/21/2021] [Indexed: 12/20/2022] Open
Abstract
Here, we identify iPLA2β as a critical regulator for p53-driven ferroptosis upon reactive oxygen species (ROS)-induced stress. The calcium-independent phospholipase iPLA2β is known to cleave acyl tails from the glycerol backbone of lipids and release oxidized fatty acids from phospholipids. We found that iPLA2β-mediated detoxification of peroxidized lipids is sufficient to suppress p53-driven ferroptosis upon ROS-induced stress, even in GPX4-null cells. Moreover, iPLA2β is overexpressed in human cancers; inhibition of endogenous iPLA2β sensitizes tumor cells to p53-driven ferroptosis and promotes p53-dependent tumor suppression in xenograft mouse models. These results demonstrate that iPLA2β acts as a major ferroptosis repressor in a GPX4-independent manner. Notably, unlike GPX4, loss of iPLA2β has no obvious effect on normal development or cell viability in normal tissues but iPLA2β plays an essential role in regulating ferroptosis upon ROS-induced stress. Thus, our study suggests that iPLA2β is a promising therapeutic target for activating ferroptosis-mediated tumor suppression without serious toxicity concerns.
Collapse
Affiliation(s)
- Delin Chen
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center,Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Bo Chu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center,Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Xin Yang
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center,Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Zhaoqi Liu
- Program for Mathematical Genomics, Departments of Systems Biology and Biomedical Informatics, Columbia University, New York, NY, 10032, USA
| | - Ying Jin
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center,Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Ning Kon
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center,Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Raul Rabadan
- Program for Mathematical Genomics, Departments of Systems Biology and Biomedical Informatics, Columbia University, New York, NY, 10032, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center,Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
20
|
Jabůrek M, Průchová P, Holendová B, Galkin A, Ježek P. Antioxidant Synergy of Mitochondrial Phospholipase PNPLA8/iPLA2γ with Fatty Acid-Conducting SLC25 Gene Family Transporters. Antioxidants (Basel) 2021; 10:antiox10050678. [PMID: 33926059 PMCID: PMC8146845 DOI: 10.3390/antiox10050678] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/13/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
Patatin-like phospholipase domain-containing protein PNPLA8, also termed Ca2+-independent phospholipase A2γ (iPLA2γ), is addressed to the mitochondrial matrix (or peroxisomes), where it may manifest its unique activity to cleave phospholipid side-chains from both sn-1 and sn-2 positions, consequently releasing either saturated or unsaturated fatty acids (FAs), including oxidized FAs. Moreover, iPLA2γ is directly stimulated by H2O2 and, hence, is activated by redox signaling or oxidative stress. This redox activation permits the antioxidant synergy with mitochondrial uncoupling proteins (UCPs) or other SLC25 mitochondrial carrier family members by FA-mediated protonophoretic activity, termed mild uncoupling, that leads to diminishing of mitochondrial superoxide formation. This mechanism allows for the maintenance of the steady-state redox status of the cell. Besides the antioxidant role, we review the relations of iPLA2γ to lipid peroxidation since iPLA2γ is alternatively activated by cardiolipin hydroperoxides and hypothetically by structural alterations of lipid bilayer due to lipid peroxidation. Other iPLA2γ roles include the remodeling of mitochondrial (or peroxisomal) membranes and the generation of specific lipid second messengers. Thus, for example, during FA β-oxidation in pancreatic β-cells, H2O2-activated iPLA2γ supplies the GPR40 metabotropic FA receptor to amplify FA-stimulated insulin secretion. Cytoprotective roles of iPLA2γ in the heart and brain are also discussed.
Collapse
Affiliation(s)
- Martin Jabůrek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1084, 14220 Prague, Czech Republic; (P.P.); (B.H.); (P.J.)
- Correspondence: ; Tel.: +420-296442789
| | - Pavla Průchová
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1084, 14220 Prague, Czech Republic; (P.P.); (B.H.); (P.J.)
| | - Blanka Holendová
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1084, 14220 Prague, Czech Republic; (P.P.); (B.H.); (P.J.)
| | - Alexander Galkin
- Department of Pediatrics, Division of Neonatology, Columbia University William Black Building, New York, NY 10032, USA;
| | - Petr Ježek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1084, 14220 Prague, Czech Republic; (P.P.); (B.H.); (P.J.)
| |
Collapse
|
21
|
Falabella M, Vernon HJ, Hanna MG, Claypool SM, Pitceathly RDS. Cardiolipin, Mitochondria, and Neurological Disease. Trends Endocrinol Metab 2021; 32:224-237. [PMID: 33640250 PMCID: PMC8277580 DOI: 10.1016/j.tem.2021.01.006] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023]
Abstract
Over the past decade, it has become clear that lipid homeostasis is central to cellular metabolism. Lipids are particularly abundant in the central nervous system (CNS) where they modulate membrane fluidity, electric signal transduction, and synaptic stabilization. Abnormal lipid profiles reported in Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and traumatic brain injury (TBI), are further support for the importance of lipid metablism in the nervous system. Cardiolipin (CL), a mitochondria-exclusive phospholipid, has recently emerged as a focus of neurodegenerative disease research. Aberrant CL content, structure, and localization are linked to impaired neurogenesis and neuronal dysfunction, contributing to aging and the pathogenesis of several neurodegenerative diseases, such as AD and PD. Furthermore, the highly tissue-specific acyl chain composition of CL confers it significant potential as a biomarker to diagnose and monitor the progression in several neurological diseases. CL also represents a potential target for pharmacological strategies aimed at treating neurodegeneration. Given the equipoise that currently exists between CL metabolism, mitochondrial function, and neurological disease, we review the role of CL in nervous system physiology and monogenic and neurodegenerative disease pathophysiology, in addition to its potential application as a biomarker and pharmacological target.
Collapse
Affiliation(s)
- Micol Falabella
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London, UK
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael G Hanna
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Steven M Claypool
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK.
| |
Collapse
|
22
|
Nessel I, Michael-Titus AT. Lipid profiling of brain tissue and blood after traumatic brain injury. Semin Cell Dev Biol 2021; 112:145-156. [DOI: 10.1016/j.semcdb.2020.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/06/2020] [Accepted: 08/08/2020] [Indexed: 11/15/2022]
|
23
|
de Oliveira LG, Angelo YDS, Iglesias AH, Peron JPS. Unraveling the Link Between Mitochondrial Dynamics and Neuroinflammation. Front Immunol 2021; 12:624919. [PMID: 33796100 PMCID: PMC8007920 DOI: 10.3389/fimmu.2021.624919] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammatory and neurodegenerative diseases are a major public health problem worldwide, especially with the increase of life-expectancy observed during the last decades. For many of these diseases, we still lack a full understanding of their etiology and pathophysiology. Nonetheless their association with mitochondrial dysfunction highlights this organelle as an important player during CNS homeostasis and disease. Markers of Parkinson (PD) and Alzheimer (AD) diseases are able to induce innate immune pathways induced by alterations in mitochondrial Ca2+ homeostasis leading to neuroinflammation. Additionally, exacerbated type I IFN responses triggered by mitochondrial DNA (mtDNA), failures in mitophagy, ER-mitochondria communication and mtROS production promote neurodegeneration. On the other hand, regulation of mitochondrial dynamics is essential for CNS health maintenance and leading to the induction of IL-10 and reduction of TNF-α secretion, increased cell viability and diminished cell injury in addition to reduced oxidative stress. Thus, although previously solely seen as power suppliers to organelles and molecular processes, it is now well established that mitochondria have many other important roles, including during immune responses. Here, we discuss the importance of these mitochondrial dynamics during neuroinflammation, and how they correlate either with the amelioration or worsening of CNS disease.
Collapse
Affiliation(s)
- Lilian Gomes de Oliveira
- Neuroimmune Interactions Laboratory, Immunology Department - Institute of Biomedical Sciences (ICB) IV, University of São Paulo (USP), São Paulo, Brazil
- Neuroimmunology of Arboviruses Laboratory, Scientific Platform Pasteur-USP, University of São Paulo (USP), São Paulo, Brazil
| | - Yan de Souza Angelo
- Neuroimmune Interactions Laboratory, Immunology Department - Institute of Biomedical Sciences (ICB) IV, University of São Paulo (USP), São Paulo, Brazil
- Neuroimmunology of Arboviruses Laboratory, Scientific Platform Pasteur-USP, University of São Paulo (USP), São Paulo, Brazil
| | - Antonio H Iglesias
- Loyola University Medical Center, Stritch School of Medicine, Loyola University Chicago, Chicago, IL, United States
| | - Jean Pierre Schatzmann Peron
- Neuroimmune Interactions Laboratory, Immunology Department - Institute of Biomedical Sciences (ICB) IV, University of São Paulo (USP), São Paulo, Brazil
- Neuroimmunology of Arboviruses Laboratory, Scientific Platform Pasteur-USP, University of São Paulo (USP), São Paulo, Brazil
- Loyola University Medical Center, Stritch School of Medicine, Loyola University Chicago, Chicago, IL, United States
| |
Collapse
|
24
|
Mowbray M, Banbury C, Rickard JJS, Davies DJ, Goldberg Oppenheimer P. Development and Characterization of a Probe Device toward Intracranial Spectroscopy of Traumatic Brain Injury. ACS Biomater Sci Eng 2021; 7:1252-1262. [PMID: 33617217 PMCID: PMC7944476 DOI: 10.1021/acsbiomaterials.0c01156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Traumatic
brain injury is a leading cause of mortality worldwide,
often affecting individuals at their most economically active yet
no primary disease-modifying interventions exist for their treatment.
Real-time direct spectroscopic examination of the brain tissue within
the context of traumatic brain injury has the potential to improve
the understanding of injury heterogeneity and subtypes, better target
management strategies and organ penetrance of pharmacological agents,
identify novel targets for intervention, and allow a clearer understanding
of fundamental biochemistry evolution. Here, a novel device is designed
and engineered, delivering Raman spectroscopy-based measurements from
the brain through clinically established cranial access techniques.
Device prototyping is undertaken within the constraints imposed by
the acquisition and site dimensions (standard intracranial access
holes, probe’s dimensions), and an artificial skull anatomical
model with cortical impact is developed. The device shows a good agreement
with the data acquired via a standard commercial
Raman, and the spectra measured are comparable in terms of quality
and detectable bands to the established traumatic brain injury model.
The developed proof-of-concept device demonstrates the feasibility
for real-time optical brain spectroscopic interface while removing
the noise of extracranial tissue and with further optimization and in vivo validation, such technology will be directly translatable
for integration into currently available standards of neurological
care.
Collapse
Affiliation(s)
- Max Mowbray
- Department of Chemical Engineering and Analytical Science, University of Manchester, The Mill, Sackwville Street, Manchester M1 3AL, U.K
| | - Carl Banbury
- School of Biochemical Engineering, EPS, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Jonathan J S Rickard
- School of Biochemical Engineering, EPS, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K.,Department of Physics, Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, U.K
| | - David J Davies
- Department of Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, National Institute for Health Research, Queen Elizabeth Hospital Birmingham, University of Birmingham, Mindelsohn Way, Birmingham B15 2TH, U.K
| | - Pola Goldberg Oppenheimer
- School of Biochemical Engineering, EPS, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K.,Healthcare Technologies Institute, Institute of Translational Medicine, Mindelsohn Way, Birmingham B15 2TH, U.K
| |
Collapse
|
25
|
Banbury C, Styles I, Eisenstein N, Zanier ER, Vegliante G, Belli A, Logan A, Goldberg Oppenheimer P. Spectroscopic detection of traumatic brain injury severity and biochemistry from the retina. BIOMEDICAL OPTICS EXPRESS 2020; 11:6249-6261. [PMID: 33282487 PMCID: PMC7687955 DOI: 10.1364/boe.399473] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/19/2020] [Accepted: 09/15/2020] [Indexed: 05/11/2023]
Abstract
Traumatic brain injury (TBI) is a major burden on healthcare services worldwide, where scientific and clinical innovation is needed to provide better understanding of biochemical damage to improve both pre-hospital assessment and intensive care monitoring. Here, we present an unconventional concept of using Raman spectroscopy to measure the biochemical response to the retina in an ex-vivo murine model of TBI. Through comparison to spectra from the brain and retina following injury, we elicit subtle spectral changes through the use of multivariate analysis, linked to a decrease in cardiolipin and indicating metabolic disruption. The ability to classify injury severity via spectra of the retina is demonstrated for severe TBI (82.0 %), moderate TBI (75.1 %) and sham groups (69.4 %). By showing that optical spectroscopy can be used to explore the eye as the window to the brain, we lay the groundwork for further exploitation of Raman spectroscopy for indirect, non-invasive assessment of brain chemistry.
Collapse
Affiliation(s)
- Carl Banbury
- School of Chemical Engineering, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Iain Styles
- Computer Science, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Neil Eisenstein
- School of Chemical Engineering, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Elisa R. Zanier
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gloria Vegliante
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Antonio Belli
- Institute of Inflammation and Ageing, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Ann Logan
- Institute of Inflammation and Ageing, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | | |
Collapse
|
26
|
Rodríguez M, Valez V, Cimarra C, Blasina F, Radi R. Hypoxic-Ischemic Encephalopathy and Mitochondrial Dysfunction: Facts, Unknowns, and Challenges. Antioxid Redox Signal 2020; 33:247-262. [PMID: 32295425 DOI: 10.1089/ars.2020.8093] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Hypoxic-ischemic events due to intrapartum complications represent the second cause of neonatal mortality and initiate an acute brain disorder known as hypoxic-ischemic encephalopathy (HIE). In HIE, the brain undergoes primary and secondary energy failure phases separated by a latent phase in which partial neuronal recovery is observed. A hypoxic-ischemic event leads to oxygen restriction causing ATP depletion, neuronal oxidative stress, and cell death. Mitochondrial dysfunction and enhanced oxidant formation in brain cells are characteristic phenomena associated with energy failure. Recent Advances: Mitochondrial sources of oxidants in neurons include complex I of the mitochondrial respiratory chain, as a key contributor to O2•- production via succinate by a reverse electron transport mechanism. The reaction of O2•- with nitric oxide (•NO) yields peroxynitrite, a mitochondrial and cellular toxin. Quantitation of the redox state of cytochrome c oxidase, through broadband near-infrared spectroscopy, represents a promising monitoring approach to evaluate mitochondrial dysfunction in vivo in humans, in conjunction with the determination of cerebral oxygenation and their correlation with the severity of brain injury. Critical Issues: The energetic failure being a key phenomenon in HIE connected with the severity of the encephalopathy, measurement of mitochondrial dysfunction in vivo provides an approach to assess evolution, prognosis, and adequate therapies. Restoration of mitochondrial redox homeostasis constitutes a key therapeutic goal. Future Directions: While hypothermia is the only currently accepted therapy in clinical management to preserve mitochondrial function, other mitochondria-targeted and/or redox-based treatments are likely to synergize to ensure further efficacy.
Collapse
Affiliation(s)
- Marianela Rodríguez
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay.,Departamento de Neonatología, Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Valeria Valez
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Carolina Cimarra
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Fernanda Blasina
- Departamento de Neonatología, Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
27
|
Lamade AM, Anthonymuthu TS, Hier ZE, Gao Y, Kagan VE, Bayır H. Mitochondrial damage & lipid signaling in traumatic brain injury. Exp Neurol 2020; 329:113307. [PMID: 32289317 DOI: 10.1016/j.expneurol.2020.113307] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria are essential for neuronal function because they serve not only to sustain energy and redox homeostasis but also are harbingers of death. A dysregulated mitochondrial network can cascade until function is irreparably lost, dooming cells. TBI is most prevalent in the young and comes at significant personal and societal costs. Traumatic brain injury (TBI) triggers a biphasic and mechanistically heterogenous response and this mechanistic heterogeneity has made the development of standardized treatments challenging. The secondary phase of TBI injury evolves over hours and days after the initial insult, providing a window of opportunity for intervention. However, no FDA approved treatment for neuroprotection after TBI currently exists. With recent advances in detection techniques, there has been increasing recognition of the significance and roles of mitochondrial redox lipid signaling in both acute and chronic central nervous system (CNS) pathologies. Oxidized lipids and their downstream products result from and contribute to TBI pathogenesis. Therapies targeting the mitochondrial lipid composition and redox state show promise in experimental TBI and warrant further exploration. In this review, we provide 1) an overview for mitochondrial redox homeostasis with emphasis on glutathione metabolism, 2) the key mechanisms of TBI mitochondrial injury, 3) the pathways of mitochondria specific phospholipid cardiolipin oxidation, and 4) review the mechanisms of mitochondria quality control in TBI with consideration of the roles lipids play in this process.
Collapse
Affiliation(s)
- Andrew M Lamade
- Department of Critical Care Medicine, Safar Center for Resuscitation Research UPMC, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Tamil S Anthonymuthu
- Department of Critical Care Medicine, Safar Center for Resuscitation Research UPMC, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary E Hier
- Department of Critical Care Medicine, Safar Center for Resuscitation Research UPMC, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Yuan Gao
- Department of Critical Care Medicine, Safar Center for Resuscitation Research UPMC, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Institute for Regenerative Medicine, IM Sechenov First Moscow State Medical University, Russian Federation
| | - Hülya Bayır
- Department of Critical Care Medicine, Safar Center for Resuscitation Research UPMC, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
28
|
Promotion of plasmalogen biosynthesis reverse lipid changes in a Barth Syndrome cell model. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158677. [PMID: 32126285 DOI: 10.1016/j.bbalip.2020.158677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/06/2020] [Accepted: 02/27/2020] [Indexed: 12/31/2022]
Abstract
In Barth syndrome (BTHS) mutations in tafazzin leads to changes in both the quantities and the molecular species of cardiolipin (CL), which are the hallmarks of BTHS. Contrary to the well-established alterations in CL associated with BTHS; recently a marked decrease in the plasmalogen levels in Barth specimens has been identified. To restore the plasmalogen levels, the present study reports the effect of promotion of plasmalogen biosynthesis on the lipidome of lymphoblasts derived from Barth patients as well as on cell viability, mitochondria biogenesis, and mitochondrial membrane potential. High resolution 31P NMR phospholipidomic analysis showed an increase in the levels of plasmenylethanolamine (the major plasmalogen in lymphoblasts), which reached values comparable to the control and a compensatory decrease in the levels of its diacyl-PE counterpart. Importantly, 31P NMR showed a significant increase in the levels of CL, while not altering the levels of monolysocardiolipin. Mass spectrometry measurements showed that the promotion of plasmalogen biosynthesis did not change the molecular species profile of targeted phospholipids. In addition, promotion of plasmalogen biosynthesis did not impact on cellular viability, although it significantly decrease mitochondria copy number and restored mitochondrial membrane potential. Overall, the results showed the efficacy of the promotion of plasmalogen biosynthesis on increasing the CL levels in a BTHS cell model and highlight the potential beneficial effect of a diet supplemented with plasmalogen precursors to BTHS patients.
Collapse
|
29
|
Kagan VE, Tyurina YY, Sun WY, Vlasova II, Dar H, Tyurin VA, Amoscato AA, Mallampalli R, van der Wel PCA, He RR, Shvedova AA, Gabrilovich DI, Bayir H. Redox phospholipidomics of enzymatically generated oxygenated phospholipids as specific signals of programmed cell death. Free Radic Biol Med 2020; 147:231-241. [PMID: 31883467 PMCID: PMC7037592 DOI: 10.1016/j.freeradbiomed.2019.12.028] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 01/16/2023]
Abstract
High fidelity and effective adaptive changes of the cell and tissue metabolism to changing environments require strict coordination of numerous biological processes. Multicellular organisms developed sophisticated signaling systems of monitoring and responding to these different contexts. Among these systems, oxygenated lipids play a significant role realized via a variety of re-programming mechanisms. Some of them are enacted as a part of pro-survival pathways that eliminate harmful or unnecessary molecules or organelles by a variety of degradation/hydrolytic reactions or specialized autophageal processes. When these "partial" intracellular measures are insufficient, the programs of cells death are triggered with the aim to remove irreparably damaged members of the multicellular community. These regulated cell death mechanisms are believed to heavily rely on signaling by a highly diversified group of molecules, oxygenated phospholipids (PLox). Out of thousands of detectable individual PLox species, redox phospholipidomics deciphered several specific molecules that seem to be diagnostic of specialized death programs. Oxygenated cardiolipins (CLs) and phosphatidylethanolamines (PEs) have been identified as predictive biomarkers of apoptosis and ferroptosis, respectively. This has led to decoding of the enzymatic mechanisms of their formation involving mitochondrial oxidation of CLs by cytochrome c and endoplasmic reticulum-associated oxidation of PE by lipoxygenases. Understanding of the specific biochemical radical-mediated mechanisms of these oxidative reactions opens new avenues for the design and search of highly specific regulators of cell death programs. This review emphasizes the usefulness of such selective lipid peroxidation mechanisms in contrast to the concept of random poorly controlled free radical reactions as instruments of non-specific damage of cells and their membranes. Detailed analysis of two specific examples of phospholipid oxidative signaling in apoptosis and ferroptosis along with their molecular mechanisms and roles in reprogramming has been presented.
Collapse
Affiliation(s)
- V E Kagan
- Center for Free Radical and Antioxidant Heath, USA; Department of Environmental and Occupational Health, University of Pittsburgh, USA; Department of Chemistry, University of Pittsburgh, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, USA; Department of Radiation Oncology, University of Pittsburgh, USA; Laboratory of Navigational Redox Lipidomics, IM Sechenov Moscow State Medical University, Moscow, Russian Federation.
| | - Y Y Tyurina
- Center for Free Radical and Antioxidant Heath, USA; Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - W Y Sun
- Center for Free Radical and Antioxidant Heath, USA; Department of Environmental and Occupational Health, University of Pittsburgh, USA; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - I I Vlasova
- Laboratory of Navigational Redox Lipidomics, IM Sechenov Moscow State Medical University, Moscow, Russian Federation
| | - H Dar
- Center for Free Radical and Antioxidant Heath, USA; Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - V A Tyurin
- Center for Free Radical and Antioxidant Heath, USA; Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - A A Amoscato
- Center for Free Radical and Antioxidant Heath, USA; Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | | | - P C A van der Wel
- Zernike Institute for Advanced Materials, University of Groningen, Groningen, the Netherlands
| | - R R He
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - A A Shvedova
- Exposure Assessment Branch, NIOSH/CDC, Morgantown, WV, USA
| | | | - H Bayir
- Center for Free Radical and Antioxidant Heath, USA; Department of Critical Care Medicine, University of Pittsburgh, USA.
| |
Collapse
|
30
|
Li L, Zhong S, Shen X, Li Q, Xu W, Tao Y, Yin H. Recent development on liquid chromatography-mass spectrometry analysis of oxidized lipids. Free Radic Biol Med 2019; 144:16-34. [PMID: 31202785 DOI: 10.1016/j.freeradbiomed.2019.06.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/21/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) in the cellular membrane can be oxidized by various enzymes or reactive oxygen species (ROS) to form many oxidized lipids. These metabolites are highly bioactive, participating in a variety of physiological and pathophysiological processes. Mass spectrometry (MS), coupled with Liquid Chromatography, has been increasingly recognized as an indispensable tool for the analysis of oxidized lipids due to its excellent sensitivity and selectivity. We will give an update on the understanding of the molecular mechanisms related to generation of various oxidized lipids and recent progress on the development of LC-MS in the detection of these bioactive lipids derived from fatty acids, cholesterol esters, and phospholipids. The purpose of this review is to provide an overview of the formation mechanisms and technological advances in LC-MS for the study of oxidized lipids in human diseases, and to shed new light on the potential of using oxidized lipids as biomarkers and mechanistic clues of pathogenesis related to lipid metabolism. The key technical problems associated with analysis of oxidized lipids and challenges in the field will also discussed.
Collapse
Affiliation(s)
- Luxiao Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China; University of Chinese Academy of Sciences, CAS, Beijing, 100049, China
| | - Shanshan Zhong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China; University of Chinese Academy of Sciences, CAS, Beijing, 100049, China
| | - Xia Shen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China; University of Chinese Academy of Sciences, CAS, Beijing, 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China
| | - Qiujing Li
- Department of Pharmacy, Zhangzhou Health Vocational College, Zhangzhou, 363000, China
| | - Wenxin Xu
- Department of Medical Technology, Zhangzhou Health Vocational College, Zhangzhou, 363000, China
| | - Yongzhen Tao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China
| | - Huiyong Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China; University of Chinese Academy of Sciences, CAS, Beijing, 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, 100000, China.
| |
Collapse
|
31
|
Anthonymuthu TS, Kenny EM, Hier ZE, Clark RSB, Kochanek PM, Kagan VE, Bayır H. Detection of brain specific cardiolipins in plasma after experimental pediatric head injury. Exp Neurol 2019; 316:63-73. [PMID: 30981805 DOI: 10.1016/j.expneurol.2019.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/14/2019] [Accepted: 04/10/2019] [Indexed: 12/20/2022]
Abstract
Cardiolipin (CL) is a mitochondria-specific phospholipid that is central to maintenance and regulation of mitochondrial bioenergetic and metabolic functions. CL molecular species display great tissue variation with brain exhibiting a distinct, highly diverse CL population. We recently showed that the appearance of unique brain-type CLs in plasma could serve as a brain-specific marker of mitochondrial/tissue injury in patients after cardiac arrest. Mitochondrial dysfunction has been increasingly implicated as a critical mechanism underlying the pathogenesis of traumatic brain injury (TBI). Therefore, we hypothesized that unique, brain-specific CL species from the injured brain are released to the peripheral circulation after TBI. To test this hypothesis, we performed a high-resolution mass spectrometry based phospholipidomics analysis of post-natal day (PND)17 rat brain and plasma after controlled cortical impact. We found a time-dependent increase in plasma CLs after TBI including the aforementioned brain-specific CL species early after injury, whereas CLs were significantly decreased in the injured brain. Compositional and quantitative correlational analysis suggested a possible release of CL into the systemic circulation following TBI. The identification of brain-type CLs in systemic circulation may indicate underlying mitochondrial dysfunction/loss after TBI. They may have potential as pharmacodynamics response biomarkers for targeted therapies.
Collapse
Affiliation(s)
- Tamil S Anthonymuthu
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Elizabeth M Kenny
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary E Hier
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Robert S B Clark
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Valerian E Kagan
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA; Laboratory of Navigational Redox Lipidomics, IM Sechenov Moscow Medical State University, Russia
| | - Hülya Bayır
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
32
|
The role of cardiolipin concentration and acyl chain composition on mitochondrial inner membrane molecular organization and function. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1039-1052. [PMID: 30951877 DOI: 10.1016/j.bbalip.2019.03.012] [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: 01/04/2019] [Revised: 03/19/2019] [Accepted: 03/30/2019] [Indexed: 12/28/2022]
Abstract
Cardiolipin (CL) is a key phospholipid of the mitochondria. A loss of CL content and remodeling of CL's acyl chains is observed in several pathologies. Strong shifts in CL concentration and acyl chain composition would presumably disrupt mitochondrial inner membrane biophysical organization. However, it remains unclear in the literature as to which is the key regulator of mitochondrial membrane biophysical properties. We review the literature to discriminate the effects of CL concentration and acyl chain composition on mitochondrial membrane organization. A widely applicable theme emerges across several pathologies, including cardiovascular diseases, diabetes, Barth syndrome, and neurodegenerative ailments. The loss of CL, often accompanied by increased levels of lyso-CLs, impairs mitochondrial inner membrane organization. Modest remodeling of CL acyl chains is not a major driver of impairments and only in cases of extreme remodeling is there an influence on membrane properties.
Collapse
|