1
|
Larrea D, Tamucci KA, Kabra K, Velasco KR, Yun TD, Pera M, Montesinos J, Agrawal RR, Paradas C, Smerdon JW, Lowry ER, Stepanova A, Yoval-Sanchez B, Galkin A, Wichterle H, Area-Gomez E. Altered mitochondria-associated ER membrane (MAM) function shifts mitochondrial metabolism in amyotrophic lateral sclerosis (ALS). Nat Commun 2025; 16:379. [PMID: 39753538 PMCID: PMC11699139 DOI: 10.1038/s41467-024-51578-1] [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: 10/27/2022] [Accepted: 08/12/2024] [Indexed: 01/06/2025] Open
Abstract
Mitochondrial function is modulated by its interaction with the endoplasmic reticulum (ER). Recent research indicates that these contacts are disrupted in familial models of amyotrophic lateral sclerosis (ALS). We report here that this impairment in the crosstalk between mitochondria and the ER impedes the use of glucose-derived pyruvate as mitochondrial fuel, causing a shift to fatty acids to sustain energy production. Over time, this deficiency alters mitochondrial electron flow and the active/dormant status of complex I in spinal cord tissues, but not in the brain. These findings suggest mitochondria-associated ER membranes (MAM domains) play a crucial role in regulating cellular glucose metabolism and that MAM dysfunction may underlie the bioenergetic deficits observed in ALS.
Collapse
Affiliation(s)
- Delfina Larrea
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Kirstin A Tamucci
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Khushbu Kabra
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Kevin R Velasco
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Taekyung D Yun
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Marta Pera
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jorge Montesinos
- Department of Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Rishi R Agrawal
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Carmen Paradas
- Department of Neurology, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - John W Smerdon
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Emily R Lowry
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna Stepanova
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Belem Yoval-Sanchez
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Alexander Galkin
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Hynek Wichterle
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Estela Area-Gomez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain.
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
2
|
Schepers M, Hendrix S, Mussen F, van Breedam E, Ponsaerts P, Lemmens S, Hellings N, Ricciarelli R, Fedele E, Bruno O, Brullo C, Prickaerts J, Van Broeckhoven J, Vanmierlo T. Amelioration of functional and histopathological consequences after spinal cord injury through phosphodiesterase 4D (PDE4D) inhibition. Neurotherapeutics 2024; 21:e00372. [PMID: 38760316 PMCID: PMC11284540 DOI: 10.1016/j.neurot.2024.e00372] [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: 12/15/2023] [Revised: 04/15/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
Spinal cord injury (SCI) is a life-changing event that severely impacts the patient's quality of life. Modulating neuroinflammation, which exacerbates the primary injury, and stimulating neuro-regenerative repair mechanisms are key strategies to improve functional recovery. Cyclic adenosine monophosphate (cAMP) is a second messenger crucially involved in both processes. Following SCI, intracellular levels of cAMP are known to decrease over time. Therefore, preventing cAMP degradation represents a promising strategy to suppress inflammation while stimulating regeneration. Intracellular cAMP levels are controlled by its hydrolyzing enzymes phosphodiesterases (PDEs). The PDE4 family is most abundantly expressed in the central nervous system (CNS) and its inhibition has been shown to be therapeutically relevant for managing SCI pathology. Unfortunately, the use of full PDE4 inhibitors at therapeutic doses is associated with severe emetic side effects, hampering their translation toward clinical applications. Therefore, in this study, we evaluated the effect of inhibiting specific PDE4 subtypes (PDE4B and PDE4D) on inflammatory and regenerative processes following SCI, as inhibitors selective for these subtypes have been demonstrated to be well-tolerated. We reveal that administration of the PDE4D inhibitor Gebr32a, even when starting 2 dpi, but not the PDE4B inhibitor A33, improved functional as well as histopathological outcomes after SCI, comparable to results obtained with the full PDE4 inhibitor roflumilast. Furthermore, using a luminescent human iPSC-derived neurospheroid model, we show that PDE4D inhibition stabilizes neural viability by preventing apoptosis and stimulating neuronal differentiation. These findings strongly suggest that specific PDE4D inhibition offers a novel therapeutic approach for SCI.
Collapse
Affiliation(s)
- Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, the Netherlands; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Sven Hendrix
- Institute for Translational Medicine, Medical School Hamburg, 20457 Hamburg, Germany
| | - Femke Mussen
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, the Netherlands; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Elise van Breedam
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610 Wilrijk, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610 Wilrijk, Belgium
| | - Stefanie Lemmens
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Niels Hellings
- University MS Centre (UMSC) Hasselt - Pelt, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Roberta Ricciarelli
- IRCCS Ospedale Policlinico San Martino, 16100 Genoa, Italy; Department of Experimental Medicine, Section of General Pathology, University of Genova, 16100 Genoa, Italy
| | - Ernesto Fedele
- IRCCS Ospedale Policlinico San Martino, 16100 Genoa, Italy; Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genoa, 16100 Genoa, Italy
| | - Olga Bruno
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, 16100 Genoa, Italy
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, 16100 Genoa, Italy
| | - Jos Prickaerts
- Peitho Translational, 6229ER Maastricht, the Netherlands
| | - Jana Van Broeckhoven
- University MS Centre (UMSC) Hasselt - Pelt, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, the Netherlands; University MS Centre (UMSC) Hasselt - Pelt, Belgium.
| |
Collapse
|
3
|
Vega-López A, Lara-Vega I, Atonal-Brioso G, Nájera-Martínez M. Neurotoxicant effects of bisphenol A, nonylphenol, and tert‑butyl phenol in the Nile tilapia (Oreochromis niloticus). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 268:106868. [PMID: 38387248 DOI: 10.1016/j.aquatox.2024.106868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/07/2024] [Accepted: 02/17/2024] [Indexed: 02/24/2024]
Abstract
Worldwide production of alkyl phenols and ethoxylated alkyl phenols is high due to their broad industrial uses. It has been widely documented that they are endocrine disruptors, and it has been suggested that they could exert neurotoxic effects. However, a lack of information about the neurotoxic effects of APs and APEs prevails. In this study, the bisphenol A (BPA), 4-nonylphenol (NP), and 3‑tert-butylphenol (tertBP) effects on brain and spinal cord of Nile tilapia exposed to environmental concentrations were evaluated by assessing acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), and carboxylesterases (CES) activities, and γ-aminobutyric acid (GABA) levels and their effects were evaluated by molecular docking. BPA and NP, tertBP behave as agonists and antagonists of AChE, BuChE, CES, and GABA, with notable differences among organs. However, none of these compounds or their metabolites interact with the enzymes' catalytic triad, suggesting an indirect alteration of enzymatic activities. While inhibiting these enzymes stand out hydrophobic interactions with the peripheral anion site, contacts with the inner face of the active site and blocking the mouth of the gorge of the active site, and steric hindrance in the enzyme pocket of glutamate decarboxylase (GAD). In contrast, inductions probably are by homotropic pseudo-cooperative phenomenon, where APEs behave as anchors favoring the active site to remain open and interactions that confer a conservative stabilization of the regulatory domain. Although the results of this study are complex, with notable differences between organs and toxicants, they are some of the first evidence of the neurotoxicity of alkylphenols and their ethoxylated derivatives.
Collapse
Affiliation(s)
- Armando Vega-López
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Toxicología Ambiental, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México City CP 07738, Mexico.
| | - Israel Lara-Vega
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Toxicología Ambiental, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México City CP 07738, Mexico
| | - Genaro Atonal-Brioso
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Toxicología Ambiental, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México City CP 07738, Mexico
| | - Minerva Nájera-Martínez
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Toxicología Ambiental, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México City CP 07738, Mexico
| |
Collapse
|
4
|
Hou B, Yin J, Liu S, Guo J, Zhang B, Zhang Z, Yang L, Tan X, Long Y, Feng S, Zhou J, Wu Y, Wang X, Han S, Wang Z, He X. Inhibiting the NLRP3 Inflammasome with MCC950 Alleviates Neurological Impairment in the Brain of EAE Mice. Mol Neurobiol 2024; 61:1318-1330. [PMID: 37702910 PMCID: PMC10896958 DOI: 10.1007/s12035-023-03618-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/27/2023] [Indexed: 09/14/2023]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by demyelination and neuronal damage. Experimental autoimmune encephalomyelitis (EAE) mice are used to model the disease progression of MS and mirror MS-like pathology. Previous researches have confirmed that inhibition of NLRP3 inflammasome significantly alleviated the severity of EAE mice and the demyelination of spinal cord, but its effect on neuronal damage and oligodendrocyte loss in the brain remains unclear. In this study, female C57BL/6 mice were immunized with MOG35-55 and PTX to establish experimental autoimmune encephalomyelitis (EAE) model. MCC950, a selective NLRP3 inflammasome inhibitor, was used to investigate the effect of NLRP3 inflammasome on the pathological changes and glial cell activation in the brain of EAE mice by immunohistochemistry. Our results demonstrated that MCC950 ameliorated the neuronal damage, demyelination, and oligodendrocyte loss in the brain of EAE mice. This protective effect of MCC950 may be attributed to its ability to suppress the activation of glial cells and prevents microglia polarization to M1 phenotype. Our work indicates that inhibition of NLRP3 inflammasome has the therapeutic effects of neuroprotection through immunomodulation and is a promising therapeutic strategy for MS.
Collapse
Affiliation(s)
- Baohua Hou
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
- Central Laboratory, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, China
| | - Jun Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Shuyan Liu
- Department of Endocrinology, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, 454000, China
| | - Jincheng Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, 454000, China
| | - Baobao Zhang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Zhenzhen Zhang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Lanping Yang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Xiying Tan
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Yijiao Long
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Sijie Feng
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Jingchun Zhou
- Beijing Bencaoyuan Pharmaceutical Co, Ltd, Beijing, 102629, China
| | - Yifan Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Xueyang Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Song Han
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Zhenhui Wang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China.
| | - Xiaohua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China.
| |
Collapse
|
5
|
Gambardella I, Worku B, Lau C, Tranbaugh RF, Tabaie S, Ivascu N, Girardi LN. Diabetes Mellitus is an Independent Predictor of Spinal Cord Injury After Descending Thoracic and Thoracoabdominal Aneurysm Repair: Maximum Likelihood Conditional Regression in a Propensity-Score Matched Cohort. Ann Surg 2023; 278:e382-e388. [PMID: 35837895 DOI: 10.1097/sla.0000000000005572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To discern the impact of diabetes mellitus (DM) on spinal cord injury (SCI) after open descending thoracic and thoracoabdominal aneurysm repair (DTAAAR). BACKGROUND Compared with euglycemia, hyperglycemia, and ketosis make neurons respectively more vulnerable and more resilient to ischemia. METHODS During the study period (1997-2021), patient who underwent DTAAAR were dichotomized according to the presence/absence of DM. The latter was investigated as predictor of our primary (SCI) and secondary [operative mortality (OM), myocardial infarction, stroke, need for tracheostomy, de novo dialysis, and survival] endpoints. Two-level risk-adjustment employed maximum likelihood conditional regression after 1:2 propensity-score matching. RESULTS DTAAAR was performed in 934 patients. Ninety-two diabetics were matched to 184 nondiabetics. All preoperative variables had a standardized mean difference <0.1 between the matched groups. Patients with DM had higher SCI (6.5% vs. 1.6%, P 0.03) and OM (14.1% vs. 6.0%, P =0.01), while the other secondary endpoints were similar between groups in the matched sample. DM was an independent predictor for SCI in the matched sample (odds ratio: 5.05, 95% confidence interval: 1.17-21.71). Matched patients with DM presented decreased survival at 1 (70.2% vs. 86.2%), 5 (50.4% vas 67.5%), 10 years (31.7% vs. 36.7%) ( P =0.03). The results are summarized in the graphical abstract. CONCLUSION DM is associated to increased OM and decreased survival, and it is an independent predictor of SCI after open DTAAAR. Strict perioperative glycemic control should be implemented, and exogenous ketones should be investigated as neuroprotective agents to reduce such adverse events.
Collapse
Affiliation(s)
- Ivancarmine Gambardella
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY
- Department of Cardiothoracic Surgery, New York Presbyterian Brooklyn Methodist Hospital, New York, NY
| | - Berhane Worku
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY
- Department of Cardiothoracic Surgery, New York Presbyterian Brooklyn Methodist Hospital, New York, NY
| | - Christopher Lau
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY
| | - Robert F Tranbaugh
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY
- Department of Cardiothoracic Surgery, New York Presbyterian Brooklyn Methodist Hospital, New York, NY
| | - Sheida Tabaie
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY
| | - Natalia Ivascu
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY
| | - Leonard N Girardi
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY
| |
Collapse
|
6
|
Expression of LIM domain-binding 3 (LDB3), a striated muscle Z-band alternatively spliced PDZ-motif protein in the nervous system. Sci Rep 2023; 13:270. [PMID: 36609526 PMCID: PMC9822979 DOI: 10.1038/s41598-023-27531-5] [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: 05/23/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
LIM domain-binding 3 (LDB3) is a member of the Enigma family of PDZ-LIM proteins. LDB3 has been reported as a striated muscle-specific Z-band alternatively spliced protein that plays an important role in mechanosensory actin cytoskeleton remodeling. This study shows that LDB3 is broadly expressed in the central and peripheral nervous system of human and mouse. LDB3 is predominantly expressed in the adult stages compared to early development and at a significantly higher level in the spinal cord than in the brain. As in skeletal muscle and heart, LDB3 is extensively alternatively spliced in the neurons. Three novel splice isoforms were identified suggesting splicing-dependent regulation of LDB3 expression in the nervous system. Expression of LDB3 in the motor cortex, cerebellum, spinal motor neuron, peripheral nerve, and neuromuscular junction in addition to skeletal muscle indicates important roles for this PDZ-LIM family protein in motor planning and execution. Moreover, expression in the hippocampal neurons suggests roles for LDB3 in learning and memory. LDB3 interactors filamin C and myotilin are also expressed in the spinal motor neuron, nerve, and neuromuscular junction, thereby providing the basis for neurogenic manifestations in myopathies associated with mutations in these so-called muscle proteins.
Collapse
|
7
|
Yu L, Fink BD, Som R, Rauckhorst AJ, Taylor EB, Sivitz WI. Metabolic clearance of oxaloacetate and mitochondrial complex II respiration: Divergent control in skeletal muscle and brown adipose tissue. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2023; 1864:148930. [PMID: 36272463 PMCID: PMC10225247 DOI: 10.1016/j.bbabio.2022.148930] [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: 03/31/2022] [Revised: 08/10/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
At low inner mitochondrial membrane potential (ΔΨ) oxaloacetate (OAA) accumulates in the organelles concurrently with decreased complex II-energized respiration. This is consistent with ΔΨ-dependent OAA inhibition of succinate dehydrogenase. To assess the metabolic importance of this process, we tested the hypothesis that perturbing metabolic clearance of OAA in complex II-energized mitochondria would alter O2 flux and, further, that this would occur in both ΔΨ and tissue-dependent fashion. We carried out respiratory and metabolite studies in skeletal muscle and interscapular brown adipose tissue (IBAT) directed at the effect of OAA transamination to aspartate (catalyzed by the mitochondrial form of glutamic-oxaloacetic transaminase, Got2) on complex II-energized respiration. Addition of low amounts of glutamate to succinate-energized mitochondria at low ΔΨ increased complex II (succinate)-energized respiration in muscle but had little effect in IBAT mitochondria. The transaminase inhibitor, aminooxyacetic acid, increased OAA concentrations and impaired succinate-energized respiration in muscle but not IBAT mitochondria at low but not high ΔΨ. Immunoblotting revealed that Got2 expression was far greater in muscle than IBAT mitochondria. Because we incidentally observed metabolism of OAA to pyruvate in IBAT mitochondria, more so than in muscle mitochondria, we also examined the expression of mitochondrial oxaloacetate decarboxylase (ODX). ODX was detected only in IBAT mitochondria. In summary, at low but not high ΔΨ, mitochondrial transamination clears OAA preventing loss of complex II respiration: a process far more active in muscle than IBAT mitochondria. We also provide evidence that OAA decarboxylation clears OAA to pyruvate in IBAT mitochondria.
Collapse
Affiliation(s)
- Liping Yu
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242, USA; Carver College of Medicine NMR Core Facility, University of Iowa, Iowa City, IA 52242, USA
| | - Brian D Fink
- Department of Internal Medicine/Endocrinology and Metabolism, University of Iowa and the Iowa City Veterans Affairs Medical Center, Iowa City, IA 52242, USA
| | - Ritu Som
- Department of Internal Medicine/Endocrinology and Metabolism, University of Iowa and the Iowa City Veterans Affairs Medical Center, Iowa City, IA 52242, USA
| | - Adam J Rauckhorst
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Eric B Taylor
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - William I Sivitz
- Department of Internal Medicine/Endocrinology and Metabolism, University of Iowa and the Iowa City Veterans Affairs Medical Center, Iowa City, IA 52242, USA.
| |
Collapse
|
8
|
Blood Metabolomics May Discriminate a Sub-Group of Patients with First Demyelinating Episode in the Context of RRMS with Increased Disability and MRI Characteristics Indicative of Poor Prognosis. Int J Mol Sci 2022; 23:ijms232314578. [PMID: 36498904 PMCID: PMC9735785 DOI: 10.3390/ijms232314578] [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: 09/30/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Biomarker research across the health-to-disease continuum is being increasingly applied. We applied blood-based metabolomics in order to identify patient clusters with a first demyelinating episode, and explored the prognostic potential of the method by thoroughly characterizing each cluster in terms of clinical, laboratory and MRI markers of established prognostic potential for Multiple Sclerosis (MS). Recruitment consisted of 11 patients with Clinically Isolated Syndrome (CIS), 37 patients with a first demyelinating episode in the context of Relapsing-Remitting MS (RRMS) and 11 control participants. Blood-based metabolomics and hierarchical clustering analysis (HCL) were applied. Constructed OPLS-DA models illustrated a discrimination between patients with CIS and the controls (p = 0.0014), as well as between patients with RRMS and the controls (p = 1 × 10−5). Hierarchical clustering analysis (HCL) for patients with RRMS identified three clusters. RRMS-patients-cluster-3 exhibited higher mean cell numbers in the Cerebro-spinal Fluid (CSF) compared to patients with CIS (18.17 ± 6.3 vs. 1.09 ± 0.41, p = 0.004). Mean glucose CSF/serum ratio and infratentorial lesion burden significantly differed across CIS- and HCL-derived RRMS-patient clusters (F = 14.95, p < 0.001 and F = 6.087, p = 0.002, respectively), mainly due to increased mean values for patients with RRMS-cluster-3. HCL discriminated a cluster of patients with a first demyelinating episode in the context of RRMS with increased disability, laboratory findings linked with increased pathology burden and MRI markers of poor prognosis.
Collapse
|
9
|
Matuz-Mares D, González-Andrade M, Araiza-Villanueva MG, Vilchis-Landeros MM, Vázquez-Meza H. Mitochondrial Calcium: Effects of Its Imbalance in Disease. Antioxidants (Basel) 2022; 11:antiox11050801. [PMID: 35624667 PMCID: PMC9138001 DOI: 10.3390/antiox11050801] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/17/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
Calcium is used in many cellular processes and is maintained within the cell as free calcium at low concentrations (approximately 100 nM), compared with extracellular (millimolar) concentrations, to avoid adverse effects such as phosphate precipitation. For this reason, cells have adapted buffering strategies by compartmentalizing calcium into mitochondria and the endoplasmic reticulum (ER). In mitochondria, the calcium concentration is in the millimolar range, as it is in the ER. Mitochondria actively contribute to buffering cellular calcium, but if matrix calcium increases beyond physiological demands, it can promote the opening of the mitochondrial permeability transition pore (mPTP) and, consequently, trigger apoptotic or necrotic cell death. The pathophysiological implications of mPTP opening in ischemia-reperfusion, liver, muscle, and lysosomal storage diseases, as well as those affecting the central nervous system, for example, Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS) have been reported. In this review, we present an updated overview of the main cellular mechanisms of mitochondrial calcium regulation. We specially focus on neurodegenerative diseases related to imbalances in calcium homeostasis and summarize some proposed therapies studied to attenuate these diseases.
Collapse
Affiliation(s)
- Deyamira Matuz-Mares
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Coyoacán, Ciudad de México 04510, Mexico; (D.M.-M.); (M.G.-A.); (M.M.V.-L.)
| | - Martin González-Andrade
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Coyoacán, Ciudad de México 04510, Mexico; (D.M.-M.); (M.G.-A.); (M.M.V.-L.)
| | | | - María Magdalena Vilchis-Landeros
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Coyoacán, Ciudad de México 04510, Mexico; (D.M.-M.); (M.G.-A.); (M.M.V.-L.)
| | - Héctor Vázquez-Meza
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Coyoacán, Ciudad de México 04510, Mexico; (D.M.-M.); (M.G.-A.); (M.M.V.-L.)
- Correspondence: ; Tel.: +52-55-5623-2168
| |
Collapse
|
10
|
Fink BD, Rauckhorst AJ, Taylor EB, Yu L, Sivitz WI. Membrane potential-dependent regulation of mitochondrial complex II by oxaloacetate in interscapular brown adipose tissue. FASEB Bioadv 2022; 4:197-210. [PMID: 35392250 PMCID: PMC8973305 DOI: 10.1096/fba.2021-00137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 11/18/2021] [Indexed: 11/11/2022] Open
Abstract
Classically, mitochondrial respiration responds to decreased membrane potential (ΔΨ) by increasing respiration. However, we found that for succinate-energized complex II respiration in skeletal muscle mitochondria (unencumbered by rotenone), low ΔΨ impairs respiration by a mechanism culminating in oxaloacetate (OAA) inhibition of succinate dehydrogenase (SDH). Here, we investigated whether this phenomenon extends to far different mitochondria of a tissue wherein ΔΨ is intrinsically low, i.e., interscapular brown adipose tissue (IBAT). Also, to advance our knowledge of the mechanism, we performed isotopomer studies of metabolite flux not done in our previous muscle studies. In additional novel work, we addressed possible ways ADP might affect the mechanism in IBAT mitochondria. UCP1 activity, and consequently ΔΨ, were perturbed both by GDP, a well-recognized potent inhibitor of UCP1 and by the chemical uncoupler carbonyl cyanide m-chlorophenyl hydrazone (FCCP). In succinate-energized mitochondria, GDP increased ΔΨ but also increased rather than decreased (as classically predicted under low ΔΨ) O2 flux. In GDP-treated mitochondria, FCCP reduced potential but also decreased respiration. Metabolite studies by NMR and flux analyses by LC-MS support a mechanism, wherein ΔΨ effects on the production of reactive oxygen alters the NADH/NAD+ ratio affecting OAA accumulation and, hence, OAA inhibition of SDH. We also found that ADP-altered complex II respiration in complex fashion probably involving decreased ΔΨ due to ATP synthesis, a GDP-like nucleotide inhibition of UCP1, and allosteric enzyme action. In summary, complex II respiration in IBAT mitochondria is regulated by UCP1-dependent ΔΨ altering substrate flow through OAA and OAA inhibition of SDH.
Collapse
Affiliation(s)
- Brian D. Fink
- Department of Internal Medicine/Endocrinology and MetabolismUniversity of Iowa and the Iowa City Veterans Affairs Medical CenterIowa CityIowaUSA
| | - Adam J. Rauckhorst
- Department of Molecular Physiology and BiophysicsUniversity of IowaIowa CityIowaUSA
| | - Eric B. Taylor
- Department of Molecular Physiology and BiophysicsUniversity of IowaIowa CityIowaUSA
| | - Liping Yu
- Department of Biochemistry and Molecular BiologyUniversity of IowaIowa CityIowaUSA
- NMR Core FacilityUniversity of IowaIowa CityIowaUSA
| | - William I. Sivitz
- Department of Internal Medicine/Endocrinology and MetabolismUniversity of Iowa and the Iowa City Veterans Affairs Medical CenterIowa CityIowaUSA
| |
Collapse
|
11
|
Tomar N, Zhang X, Kandel SM, Sadri S, Yang C, Liang M, Audi SH, Cowley AW, Dash RK. Substrate-dependent differential regulation of mitochondrial bioenergetics in the heart and kidney cortex and outer medulla. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148518. [PMID: 34864090 PMCID: PMC8957717 DOI: 10.1016/j.bbabio.2021.148518] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/29/2021] [Accepted: 11/20/2021] [Indexed: 05/05/2023]
Abstract
The kinetics and efficiency of mitochondrial oxidative phosphorylation (OxPhos) can depend on the choice of respiratory substrates. Furthermore, potential differences in this substrate dependency among different tissues are not well-understood. Here, we determined the effects of different substrates on the kinetics and efficiency of OxPhos in isolated mitochondria from the heart and kidney cortex and outer medulla (OM) of Sprague-Dawley rats. The substrates were pyruvate+malate, glutamate+malate, palmitoyl-carnitine+malate, alpha-ketoglutarate+malate, and succinate±rotenone at saturating concentrations. The kinetics of OxPhos were interrogated by measuring mitochondrial bioenergetics under different ADP perturbations. Results show that the kinetics and efficiency of OxPhos are highly dependent on the substrates used, and this dependency is distinctly different between heart and kidney. Heart mitochondria showed higher respiratory rates and OxPhos efficiencies for all substrates in comparison to kidney mitochondria. Cortex mitochondria respiratory rates were higher than OM mitochondria, but OM mitochondria OxPhos efficiencies were higher than cortex mitochondria. State 3 respiration was low in heart mitochondria with succinate but increased significantly in the presence of rotenone, unlike kidney mitochondria. Similar differences were observed in mitochondrial membrane potential. Differences in H2O2 emission in the presence of succinate±rotenone were observed in heart mitochondria and to a lesser extent in OM mitochondria, but not in cortex mitochondria. Bioenergetics and H2O2 emission data with succinate±rotenone indicate that oxaloacetate accumulation and reverse electron transfer may play a more prominent regulatory role in heart mitochondria than kidney mitochondria. These studies provide novel quantitative data demonstrating that the choice of respiratory substrates affects mitochondrial responses in a tissue-specific manner.
Collapse
Affiliation(s)
- Namrata Tomar
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Xiao Zhang
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Sunil M Kandel
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Shima Sadri
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Said H Audi
- Department of Biomedical Engineering, Marquette University, Milwaukee WI-53223, United States of America
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee WI-53226, United States of America.
| | - Ranjan K Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee WI-53226, United States of America.
| |
Collapse
|
12
|
Fu J, Xu W, Zhang Y, Sun H, Zhao J. Luteolin Modulates the NF-E2-Related Factor 2/Glutamate-Cysteine Ligase Pathway in Rats with Spinal Cord Injury. J Med Food 2021; 24:218-225. [PMID: 33739882 DOI: 10.1089/jmf.2020.4764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Spinal cord ischemia-reperfusion injury (SCII) easily causes unalterable neurological deficits. We previously demonstrated that the flavonoid luteolin (LU) has strong antioxidant, anti-inflammatory, and other neuroprotective efficacies against SCII. In our current study, we examined the contributions of the NF-E2-related factor 2 (Nrf2)/glutamate-cysteine ligase (GCL) pathway to LU-mediated neuroprotection in the transient abdominal aorta occlusion rat model of SCII. Rats were divided into four groups: Sham surgery, SCII alone, SCII plus LU pretreatment (SCII + LU), and SCII plus cotreatment with LU and the Nrf2 inhibitor ML385 (SCII + LU + ML385). The Basso-Beattie-Bresnahan (BBB) scale was used to assess neurological function, hematoxylin and eosin staining to evaluate pathological change to the spinal cord, and enzyme-linked immunosorbent assay to measure tissue markers of oxidative stress and inflammation induced by SCII. Mitochondrial injury and apoptosis were examined by flow cytometry and expression levels of Nrf2, GCL catalytic subunit (GCLc), and GCL modifier subunit (GCLm) by real-time quantitative polymerase chain reaction. LU pretreatment significantly enhanced recovery of motor function as evidenced by the BBB score and attenuated the pathological damage. Furthermore, LU effectively enhanced the antioxidative activity, alleviated mitochondrial swelling, decreased the expression levels of several proinflammatory cytokines after SCII, and significantly upregulated Nrf2, GCLc, and GCLm expression levels. Cotreatment with ML385 reversed all these protective effects of LU except the anti-inflammatory response. Collectively, these findings indicate that the neuroprotective efficacy of LU depends on suppression of oxidative stress and preservation of mitochondrial function through signaling pathways involving Nrf2 activation and downstream gene expression.
Collapse
Affiliation(s)
- Jian Fu
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wei Xu
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yongzhe Zhang
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haibin Sun
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianhui Zhao
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
13
|
Gambardella I, Ascione R, D'Agostino DP, Ari C, Worku B, Tranbaugh RF, Ivascu N, Villena-Vargas J, Girardi LN. Systematic Review - Neuroprotection of ketosis in acute injury of the mammalian central nervous system: A meta-analysis. J Neurochem 2021; 158:105-118. [PMID: 33675563 DOI: 10.1111/jnc.15341] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/27/2021] [Indexed: 11/27/2022]
Abstract
To evaluate the neuroprotection exerted by ketosis against acute damage of the mammalian central nervous system (CNS). Search engines were interrogated to identify experimental studies comparing the mitigating effect of ketosis (intervention) versus non-ketosis (control) on acute CNS damage. Primary endpoint was a reduction in mortality. Secondary endpoints were a reduction in neuronal damage and dysfunction, and an 'aggregated advantage' (composite of all primary and secondary endpoints). Hedges' g was the effect measure. Subgroup analyses evaluated the modulatory effect of age, insult type, and injury site. Meta-regression evaluated timing, type, and magnitude of intervention as predictors of neuroprotection. The selected publications were 49 experimental murine studies (period 1979-2020). The intervention reduced mortality (g 2.45, SE 0.48, p < .01), neuronal damage (g 1.96, SE 0.23, p < .01) and dysfunction (g 0.99, SE 0.10, p < .01). Reduction of mortality was particularly pronounced in the adult subgroup (g 2.71, SE 0.57, p < .01). The aggregated advantage of ketosis was stronger in the pediatric (g 3.98, SE 0.71, p < .01), brain (g 1.96, SE 0.18, p < .01), and ischemic insult (g 2.20, SE 0.23, p < .01) subgroups. Only the magnitude of intervention was a predictor of neuroprotection (g 0.07, SE 0.03, p 0.01 per every mmol/L increase in ketone levels). Ketosis exerts a potent neuroprotection against acute damage to the mammalian CNS in terms of reduction of mortality, of neuronal damage and dysfunction. Hematic levels of ketones are directly proportional to the effect size of neuroprotection.
Collapse
Affiliation(s)
| | - Raimondo Ascione
- Bristol Translational Biomedical Research Centre, University of Bristol, Bristol, UK
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Csilla Ari
- Behavioral Neuroscience Research Laboratory, Department of Psychology, University of South Florida, Tampa, FL, USA
| | - Berhane Worku
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Robert F Tranbaugh
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Natalia Ivascu
- Department of Anesthesia, Weill Cornell Medicine, New York, NY, USA
| | | | - Leonard N Girardi
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
14
|
Serial Systemic Injections of Endotoxin (LPS) Elicit Neuroprotective Spinal Cord Microglia through IL-1-Dependent Cross Talk with Endothelial Cells. J Neurosci 2020; 40:9103-9120. [PMID: 33051350 DOI: 10.1523/jneurosci.0131-20.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Microglia are dynamic immunosurveillance cells in the CNS. Whether microglia are protective or pathologic is context dependent; the outcome varies as a function of time relative to the stimulus, activation state of neighboring cells in the microenvironment or within progression of a particular disease. Although brain microglia can be "primed" using bacterial lipopolysaccharide (LPS)/endotoxin, it is unknown whether LPS delivered systemically can also induce neuroprotective microglia in the spinal cord. Here, we show that serial systemic injections of LPS (1 mg/kg, i.p., daily) for 4 consecutive days (LPSx4) consistently elicit a reactive spinal cord microglia response marked by dramatic morphologic changes, increased production of IL-1, and enhanced proliferation without triggering leukocyte recruitment or overt neuropathology. Following LPSx4, reactive microglia frequently contact spinal cord endothelial cells. Targeted ablation or selective expression of IL-1 and IL-1 receptor (IL-1R) in either microglia or endothelia reveal that IL-1-dependent signaling between these cells mediates microglia activation. Using a mouse model of ischemic spinal cord injury in male and female mice, we show that preoperative LPSx4 provides complete protection from ischemia-induced neuron loss and hindlimb paralysis. Neuroprotection is partly reversed by either pharmacological elimination of microglia or selective removal of IL-1R in microglia or endothelia. These data indicate that spinal cord microglia are amenable to therapeutic reprogramming via systemic manipulation and that this potential can be harnessed to protect the spinal cord from injury.SIGNIFICANCE STATEMENT Data in this report indicate that a neuroprotective spinal cord microglia response can be triggered by daily systemic injections of LPS over a period of 4 d (LPSx4). The LPSx4 regimen induces morphologic transformation and enhances proliferation of spinal cord microglia without causing neuropathology. Using advanced transgenic mouse technology, we show that IL-1-dependent microglia-endothelia cross talk is necessary for eliciting this spinal cord microglia phenotype and also for conferring optimal protection to spinal motor neurons from ischemic spinal cord injury (ISCI). Collectively, these novel data show that it is possible to consistently elicit spinal cord microglia via systemic delivery of inflammogens to achieve a therapeutically effective neuroprotective response against ISCI.
Collapse
|
15
|
Filadi R, Pizzo P. Mitochondrial calcium handling and neurodegeneration: when a good signal goes wrong. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
16
|
Fu J, Sun H, Wei H, Dong M, Zhang Y, Xu W, Fang Y, Zhao J. Astaxanthin alleviates spinal cord ischemia-reperfusion injury via activation of PI3K/Akt/GSK-3β pathway in rats. J Orthop Surg Res 2020; 15:275. [PMID: 32703256 PMCID: PMC7376638 DOI: 10.1186/s13018-020-01790-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Ischemia-reperfusion injury of the spinal cord (SCII) often leads to unalterable neurological deficits, which may be associated with apoptosis induced by oxidative stress and inflammation. Astaxanthin (AST) is a strong antioxidant and anti-inflammatory agent with multitarget neuroprotective effects. This study aimed to investigate the potential therapeutic effects of AST for SCII and the molecular mechanism. METHODS Rat models of SCII with abdominal aortic occlusion for 40 min were carried out to investigate the effects of AST on the recovery of SCII. Tarlov's scores were used to assess the neuronal function; HE and TUNEL staining were used to observe the pathological morphology of lesions. Neuron oxidative stress and inflammation were measured using commercial detection kits. Flow cytometry was conducted to assess the mitochondrial swelling degree. Besides, Western blot assay was used to detect the expression of PI3K/Akt/GSK-3β pathway-related proteins, as well as NOX2 and NLRP3 proteins. RESULTS The results demonstrated that AST pretreatment promoted the hind limb motor function recovery and alleviated the pathological damage induced by SCII. Moreover, AST significantly enhanced the antioxidative stress response and attenuated mitochondrial swelling. However, AST pretreatment hardly inhibited the levels of proinflammatory cytokines after SCII. Most importantly, AST activated p-Akt and p-GSK-3β expression levels. Meanwhile, cotreatment with LY294002 (a PI3K inhibitor) was found to abolish the above protective effects observed with the AST pretreatment. CONCLUSION Overall, these results suggest that AST pretreatment not only mitigates pathological tissue damage but also effectively improves neural functional recovery following SCII, primarily by alleviating oxidative stress but not inhibiting inflammation. A possible underlying molecular mechanism of AST may be mainly attributed to the activation of PI3K/Akt/GSK-3β pathway.
Collapse
Affiliation(s)
- Jian Fu
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Haibin Sun
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Haofei Wei
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Mingjie Dong
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Yongzhe Zhang
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Wei Xu
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Yanwei Fang
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Jianhui Zhao
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
17
|
Bonomo R, Cavaletti G, Skene DJ. Metabolomics markers in Neurology: current knowledge and future perspectives for therapeutic targeting. Expert Rev Neurother 2020; 20:725-738. [PMID: 32538242 DOI: 10.1080/14737175.2020.1782746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Metabolomics is an emerging approach providing new insights into the metabolic changes and underlying mechanisms involved in the pathogenesis of neurological disorders. AREAS COVERED Here, the authors present an overview of the current knowledge of metabolic profiling (metabolomics) to provide critical insight on the role of biochemical markers and metabolic alterations in neurological diseases. EXPERT OPINION Elucidation of characteristic metabolic alterations in neurological disorders is crucial for a better understanding of their pathogenesis, and for identifying potential biomarkers and drug targets. Nevertheless, discrepancies in diagnostic criteria, sample handling protocols, and analytical methods still affect the generalizability of current study results.
Collapse
Affiliation(s)
- Roberta Bonomo
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca , Monza, Italy.,Chronobiology, Faculty of Health and Medical Sciences, University of Surrey , Guildford, UK
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca , Monza, Italy
| | - Debra J Skene
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey , Guildford, UK
| |
Collapse
|
18
|
Bursch F, Kalmbach N, Naujock M, Staege S, Eggenschwiler R, Abo-Rady M, Japtok J, Guo W, Hensel N, Reinhardt P, Boeckers TM, Cantz T, Sterneckert J, Van Den Bosch L, Hermann A, Petri S, Wegner F. Altered calcium dynamics and glutamate receptor properties in iPSC-derived motor neurons from ALS patients with C9orf72, FUS, SOD1 or TDP43 mutations. Hum Mol Genet 2020; 28:2835-2850. [PMID: 31108504 DOI: 10.1093/hmg/ddz107] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/02/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022] Open
Abstract
The fatal neurodegenerative disease amyotrophic lateral sclerosis (ALS) is characterized by a profound loss of motor neurons (MNs). Until now only riluzole minimally extends life expectancy in ALS, presumably by inhibiting glutamatergic neurotransmission and calcium overload of MNs. Therefore, the aim of this study was to investigate the glutamate receptor properties and key aspects of intracellular calcium dynamics in induced pluripotent stem cell (iPSC)-derived MNs from ALS patients with C9orf72 (n = 4 cell lines), fused in sarcoma (FUS) (n = 9), superoxide dismutase 1 (SOD1) (n = 3) or transactive response DNA-binding protein 43 (TDP43) (n = 3) mutations as well as healthy (n = 7 cell lines) and isogenic controls (n = 3). Using calcium imaging, we most frequently observed spontaneous transients in mutant C9orf72 MNs. Basal intracellular calcium levels and α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA)-induced signal amplitudes were elevated in mutant TDP43 MNs. Besides, a majority of mutant TDP43 MNs responded to 3.5-dihydroxyphenylglycine as metabotropic glutamate receptor agonist. Quantitative real-time PCR demonstrated significantly increased expression levels of AMPA and kainate receptors in mutant FUS cells compared to healthy and isogenic controls. Furthermore, the expression of kainate receptors and voltage gated calcium channels in mutant C9orf72 MNs as well as metabotropic glutamate receptors in mutant SOD1 cells was markedly elevated compared to controls. Our data of iPSC-derived MNs from familial ALS patients revealed several mutation-specific alterations in glutamate receptor properties and calcium dynamics that could play a role in ALS pathogenesis and may lead to future translational strategies with individual stratification of neuroprotective ALS treatments.
Collapse
Affiliation(s)
- Franziska Bursch
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Norman Kalmbach
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Maximilian Naujock
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Selma Staege
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Reto Eggenschwiler
- Research Group Translational Hepatology and Stem Cell Biology, Cluster of Excellence REBIRTH, Hannover Medical School, 30625 Hannover, Germany
| | | | - Julia Japtok
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Wenting Guo
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, BE-3000 Leuven, Belgium.,Laboratory of Neurobiology, VIB-Center for Brain & Disease Research, BE-3000 Leuven, Belgium
| | - Niko Hensel
- Institute of Neuroanatomy, Hannover Medical School, 30625 Hanover, Germany
| | | | - Tobias M Boeckers
- Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Tobias Cantz
- Research Group Translational Hepatology and Stem Cell Biology, Cluster of Excellence REBIRTH, Hannover Medical School, 30625 Hannover, Germany
| | | | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, BE-3000 Leuven, Belgium.,Laboratory of Neurobiology, VIB-Center for Brain & Disease Research, BE-3000 Leuven, Belgium
| | - Andreas Hermann
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| |
Collapse
|
19
|
Fink BD, Yu L, Sivitz WI. Modulation of complex II-energized respiration in muscle, heart, and brown adipose mitochondria by oxaloacetate and complex I electron flow. FASEB J 2019; 33:11696-11705. [PMID: 31361970 PMCID: PMC6902704 DOI: 10.1096/fj.201900690r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/01/2019] [Indexed: 01/10/2023]
Abstract
We recently reported that membrane potential (ΔΨ) primarily determines the relationship of complex II-supported respiration by isolated skeletal muscle mitochondria to ADP concentrations. We observed that O2 flux peaked at low ADP concentration ([ADP]) (high ΔΨ) before declining at higher [ADP] (low ΔΨ). The decline resulted from oxaloacetate (OAA) accumulation and inhibition of succinate dehydrogenase. This prompted us to question the effect of incremental [ADP] on respiration in interscapular brown adipose tissue (IBAT) mitochondria, wherein ΔΨ is intrinsically low because of uncoupling protein 1 (UCP1). We found that succinate-energized IBAT mitochondria, even in the absence of ADP, accumulate OAA and manifest limited respiration, similar to muscle mitochondria at high [ADP]. This could be prevented by guanosine 5'-diphosphate inhibition of UCP1. NAD+ cycling with NADH requires complex I electron flow and is needed to form OAA. Therefore, to assess the role of electron transit, we perturbed flow using a small molecule, N1-(3-acetamidophenyl)-N2-(2-(4-methyl-2-(p-tolyl)thiazol-5-yl)ethyl)oxalamide. We observed decreased OAA, increased NADH/NAD+, and increased succinate-supported mitochondrial respiration under conditions of low ΔΨ (IBAT) but not high ΔΨ (heart). In summary, complex II-energized respiration in IBAT mitochondria is tempered by complex I-derived OAA in a manner dependent on UCP1. These dynamics depend on electron transit in complex I.-Fink, B. D., Yu, L., Sivitz, W. I. Modulation of complex II-energized respiration in muscle, heart, and brown adipose mitochondria by oxaloacetate and complex I electron flow.
Collapse
Affiliation(s)
- Brian D. Fink
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa–Iowa City Veterans Affairs Health Care System, Iowa City, Iowa, USA
| | - Liping Yu
- Department of Biochemistry, University of Iowa–Iowa City Veterans Affairs Health Care System, Iowa City, Iowa, USA
- NMR Core Facility, University of Iowa–Iowa City Veterans Affairs Health Care System, Iowa City, Iowa, USA
| | - William I. Sivitz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa–Iowa City Veterans Affairs Health Care System, Iowa City, Iowa, USA
| |
Collapse
|
20
|
Garland J, Olds K, Rousseau G, Palmiere C, Ondruschka B, Kesha K, Glenn C, Morrow P, Stables S, Tse R. Using vitreous humour and cerebrospinal fluid electrolytes in estimating post-mortem interval - an exploratory study. AUST J FORENSIC SCI 2019. [DOI: 10.1080/00450618.2019.1642956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jack Garland
- Forensic and Analytical Science Service, NSW Health Pathology, Lidcombe, Australia
| | - Kelly Olds
- School of Medicine, University of Notre Dame, Fremantle, Australia
| | - Guillaume Rousseau
- Department of Biochemistry and Genetics, University Hospital of Angers, Angers, France
| | - Cristian Palmiere
- CURML, University Center of Legal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | | | - K. Kesha
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Charley Glenn
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - P. Morrow
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - S. Stables
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - R. Tse
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
21
|
Garland J, Philcox W, McCarthy S, Kesha K, Lam L, Spark A, Palmiere C, Elstub H, Cala A, Stables S, Tse R. Post-mortem biochemistry differences between vitreous humour and cerebrospinal fluid. AUST J FORENSIC SCI 2019. [DOI: 10.1080/00450618.2019.1597920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- J. Garland
- Hornsby Ku-Ring-Gai Hospital, Hornsby, New South Wales, Australia
| | - W. Philcox
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sinead McCarthy
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - K. Kesha
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - L. Lam
- Department of Biochemistry, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - A. Spark
- Wellington Hospital Mortuary, Wellington, New Zealand
| | - C. Palmiere
- CURML, University Center of Legal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - H. Elstub
- Department of Forensic Medicine, Newcastle, Forensic & Analytical Science Service (FASS), NSW Health Pathology, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - A.D. Cala
- Department of Forensic Medicine, Newcastle, Forensic & Analytical Science Service (FASS), NSW Health Pathology, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - S. Stables
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - R. Tse
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
22
|
Upadhyay A, Hosseinibarkooie S, Schneider S, Kaczmarek A, Torres-Benito L, Mendoza-Ferreira N, Overhoff M, Rombo R, Grysko V, Kye MJ, Kononenko NL, Wirth B. Neurocalcin Delta Knockout Impairs Adult Neurogenesis Whereas Half Reduction Is Not Pathological. Front Mol Neurosci 2019; 12:19. [PMID: 30853885 PMCID: PMC6396726 DOI: 10.3389/fnmol.2019.00019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 01/21/2019] [Indexed: 12/22/2022] Open
Abstract
Neurocalcin delta (NCALD) is a brain-enriched neuronal calcium sensor and its reduction acts protective against spinal muscular atrophy (SMA). However, the physiological function of NCALD and implications of NCALD reduction are still elusive. Here, we analyzed the ubiquitous Ncald knockout in homozygous (NcaldKO/KO) and heterozygous (NcaldKO/WT) mice to unravel the physiological role of NCALD in the brain and to study whether 50% NCALD reduction is a safe option for SMA therapy. We found that NcaldKO/KO but not NcaldKO/WT mice exhibit significant changes in the hippocampal morphology, likely due to impaired generation and migration of newborn neurons in the dentate gyrus (DG). To understand the mechanism behind, we studied the NCALD interactome and identified mitogen-activated protein kinase kinase kinase 10 (MAP3K10) as a novel NCALD interacting partner. MAP3K10 is an upstream activating kinase of c-Jun N-terminal kinase (JNK), which regulates adult neurogenesis. Strikingly, the JNK activation was significantly upregulated in the NcaldKO/KO brains. Contrary, neither adult neurogenesis nor JNK activation were altered by heterozygous Ncald deletion. Taken together, our study identifies a novel link between NCALD and adult neurogenesis in the hippocampus, possibly via a MAP3K10-JNK pathway and emphasizes the safety of using NCALD reduction as a therapeutic option for SMA.
Collapse
Affiliation(s)
- Aaradhita Upadhyay
- Institute of Human Genetics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Seyyedmohsen Hosseinibarkooie
- Institute of Human Genetics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Svenja Schneider
- Institute of Human Genetics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Anna Kaczmarek
- Institute of Human Genetics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Laura Torres-Benito
- Institute of Human Genetics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Natalia Mendoza-Ferreira
- Institute of Human Genetics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Melina Overhoff
- Institute for Genetics, University of Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Roman Rombo
- Institute of Human Genetics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Vanessa Grysko
- Institute of Human Genetics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Min Jeong Kye
- Institute of Human Genetics, University of Cologne, Cologne, Germany
| | - Natalia L Kononenko
- Institute for Genetics, University of Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany.,Center for Rare Diseases Cologne, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
23
|
Garland J, Philcox W, Kesha K, Morrow P, Lam L, Spark A, Palmiere C, Elstub H, Cala AD, Stables S, Tse R. Differences in Sampling Site on Postmortem Cerebrospinal Fluid Biochemistry. ACTA ACUST UNITED AC 2018; 39:304-308. [DOI: 10.1097/paf.0000000000000420] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
24
|
Biochemical Differences Between Vitreous Humor and Cerebral Spinal Fluid in a Death From Diabetic Ketoacidosis. Am J Forensic Med Pathol 2018; 40:188-191. [PMID: 30475235 DOI: 10.1097/paf.0000000000000450] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Biochemical analysis of cerebrospinal fluid (CSF) and vitreous humor (VH) forms an important diagnostic ancillary test at autopsy. Cerebrospinal fluid can be sampled from the ventricular system (VA) and from lumbar puncture (LP), whereas VH can be sampled from the orbits. Biochemical electrolytes seem to vary between VH and CSF collected from different sites, but whether there is any difference in glucose and β-hydroxybutyrate is unclear. We present a case report of a 21-year-old man who died of diabetic ketoacidosis confirmed at autopsy. Ventricular system, LP, and VH were biochemically analyzed and showed that glucose was highest in VH (41 mmol/L) and was 6 and 8 mmol/L higher than CSF in the LP and VA, respectively. β-Hydroxybutyrate was also highest in VH (20 mmol/L) and was 5 and 6 mmol/L higher than LP and VA, respectively. Our findings suggest that postmortem CSF glucose and β-hydroxybutyrate may not truly reflect that of VH and vary between CSF sampling sites.
Collapse
|
25
|
Fink BD, Bai F, Yu L, Sheldon RD, Sharma A, Taylor EB, Sivitz WI. Oxaloacetic acid mediates ADP-dependent inhibition of mitochondrial complex II-driven respiration. J Biol Chem 2018; 293:19932-19941. [PMID: 30385511 DOI: 10.1074/jbc.ra118.005144] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/30/2018] [Indexed: 01/01/2023] Open
Abstract
We recently reported a previously unrecognized mitochondrial respiratory phenomenon. When [ADP] was held constant ("clamped") at sequentially increasing concentrations in succinate-energized muscle mitochondria in the absence of rotenone (commonly used to block complex I), we observed a biphasic, increasing then decreasing, respiratory response. Here we investigated the mechanism. We confirmed decades-old reports that oxaloacetate (OAA) inhibits succinate dehydrogenase (SDH). We then used an NMR method to assess OAA concentrations (known as difficult to measure by MS) as well as those of malate, fumarate, and citrate in isolated succinate-respiring mitochondria. When these mitochondria were incubated at varying clamped ADP concentrations, respiration increased at low [ADP] as expected given the concurrent reduction in membrane potential. With further increments in [ADP], respiration decreased associated with accumulation of OAA. Moreover, a low pyruvate concentration, that alone was not enough to drive respiration, was sufficient to metabolize OAA to citrate and completely reverse the loss of succinate-supported respiration at high [ADP]. Further, chemical or genetic inhibition of pyruvate uptake prevented OAA clearance and preserved respiration. In addition, we measured the effects of incremental [ADP] on NADH, superoxide, and H2O2 (a marker of reverse electron transport from complex II to I). In summary, our findings, taken together, support a mechanism (detailed within) wherein succinate-energized respiration as a function of increasing [ADP] is initially increased by [ADP]-dependent effects on membrane potential but subsequently decreased at higher [ADP] by inhibition of succinate dehydrogenase by OAA. The physiologic relevance is discussed.
Collapse
Affiliation(s)
- Brian D Fink
- From the Department of Internal Medicine/Endocrinology and Metabolism
| | - Fan Bai
- From the Department of Internal Medicine/Endocrinology and Metabolism
| | - Liping Yu
- Department of Biochemistry, and.,NMR Core Facility, University of Iowa and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52242
| | | | | | | | - William I Sivitz
- From the Department of Internal Medicine/Endocrinology and Metabolism,
| |
Collapse
|
26
|
Gorter RP, Nutma E, Jahrei M, de Jonge JC, Quinlan RA, van der Valk P, van Noort JM, Baron W, Amor S. Heat shock proteins are differentially expressed in brain and spinal cord: implications for multiple sclerosis. Clin Exp Immunol 2018; 194:137-152. [PMID: 30014472 PMCID: PMC6194336 DOI: 10.1111/cei.13186] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2018] [Indexed: 01/10/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease characterized by demyelination, inflammation and neurodegeneration throughout the central nervous system. Although spinal cord pathology is an important factor contributing to disease progression, few studies have examined MS lesions in the spinal cord and how they differ from brain lesions. In this study we have compared brain and spinal cord white (WM) and grey (GM) matter from MS and control tissues, focusing on small heat shock proteins (HSPB) and HSP16.2. Western blotting was used to examine protein levels of HSPB1, HSPB5, HSPB6, HSPB8 and HSP16.2 in brain and spinal cord from MS and age-matched non-neurological controls. Immunohistochemistry was used to examine expression of the HSPs in MS spinal cord lesions and controls. Expression levels were quantified using ImageJ. Western blotting revealed significantly higher levels of HSPB1, HSPB6 and HSPB8 in MS and control spinal cord compared to brain tissues. No differences in HSPB5 and HSP16.2 protein levels were observed, although HSPB5 protein levels were higher in brain WM versus GM. In MS spinal cord lesions, increased HSPB1 and HSPB5 expression was observed in astrocytes, and increased neuronal expression of HSP16.2 was observed in normal-appearing GM and type 1 GM lesions. The high constitutive expression of several HSPBs in spinal cord and increased expression of HSPBs and HSP16.2 in MS illustrate differences between brain and spinal cord in health and upon demyelination. Regional differences in HSP expression may reflect differences in astrocyte cytoskeleton composition and influence inflammation, possibly affecting the effectiveness of pharmacological agents.
Collapse
Affiliation(s)
- R. P. Gorter
- Pathology DepartmentAmsterdam UMC, VUMCGroningenUK
| | - E. Nutma
- Pathology DepartmentAmsterdam UMC, VUMCGroningenUK
| | - M.‐C. Jahrei
- Pathology DepartmentAmsterdam UMC, VUMCGroningenUK
| | - J. C. de Jonge
- Department of Cell BiologyUniversity of Groningen, University Medical Center GroningenGroningenUK
| | - R. A Quinlan
- Department of BiosciencesDurham UniversityDurhamUK
| | | | | | - W. Baron
- Department of Cell BiologyUniversity of Groningen, University Medical Center GroningenGroningenUK
| | - S. Amor
- Pathology DepartmentAmsterdam UMC, VUMCGroningenUK
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
27
|
Martin LJ, Chang Q. DNA Damage Response and Repair, DNA Methylation, and Cell Death in Human Neurons and Experimental Animal Neurons Are Different. J Neuropathol Exp Neurol 2018; 77:636-655. [PMID: 29788379 PMCID: PMC6005106 DOI: 10.1093/jnen/nly040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neurological disorders affecting individuals in infancy to old age elude interventions for meaningful protection against neurodegeneration, and preclinical work has not translated to humans. We studied human neuron responses to injury and death stimuli compared to those of animal neurons in culture under similar settings of insult (excitotoxicity, oxidative stress, and DNA damage). Human neurons were differentiated from a cortical neuron cell line and the embryonic stem cell-derived H9 line. Mouse neurons were differentiated from forebrain neural stem cells and embryonic cerebral cortex; pig neurons were derived from forebrain neural stem cells. Mitochondrial morphology was different in human and mouse neurons. Human and mouse neurons challenged with DNA-damaging agent camptothecin showed different chromatin condensation, cell death, and DNA damage sensor activation. DNA damage accumulation and repair kinetics differed among human, mouse, and pig neurons. Promoter CpG island methylation microarrays showed significant differential DNA methylation in human and mouse neurons after injury. Therefore, DNA damage response, DNA repair, DNA methylation, and autonomous cell death mechanisms in human neurons and experimental animal neurons are different.
Collapse
Affiliation(s)
- Lee J Martin
- Department of Pathology, Division of Neuropathology
- Pathobiology Graduate Training Program
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qing Chang
- Department of Pathology, Division of Neuropathology
| |
Collapse
|
28
|
Panov A, Orynbayeva Z. Determination of mitochondrial metabolic phenotype through investigation of the intrinsic inhibition of succinate dehydrogenase. Anal Biochem 2017; 552:30-37. [PMID: 29042133 DOI: 10.1016/j.ab.2017.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 11/25/2022]
Abstract
Many diseases are accompanied by systemic or organ metabolic abnormalities. Therefore, investigation of the roles of mitochondrial dysfunction in the pathogenesis of major diseases requires a methodology that reflects the characteristics of mitochondrial metabolism particular for the organ of origin. We provide evidence that for brain and heart mitochondria the intrinsic inhibition of succinate dehydrogenase (SDH) is a key mechanism for attenuation of mitochondrial respiration and energy production in response to the organ's energy needs. This mechanism also serves to minimize the production of reactive oxygen species when the organ is at rest. Changes in the organ's workloads are accompanied by changes in metabolites that are used by mitochondria as substrates and for modification of energy production at the SDH level. Measurement of the respiratory activity of mitochondria with various substrates and substrate mixtures and use of bovine serum albumin as an SDH inhibitor will be useful for evaluation of metabolic phenotype at the mitochondrial level.
Collapse
Affiliation(s)
- Alexander Panov
- Institute of Molecular Biology and Biophysics, Russian Academy of Sciences, Siberian Branch, Novosibirsk 630117, Russia.
| | - Zulfiya Orynbayeva
- Department of Surgery, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
29
|
Bai F, Fink BD, Yu L, Sivitz WI. Voltage-Dependent Regulation of Complex II Energized Mitochondrial Oxygen Flux. PLoS One 2016; 11:e0154982. [PMID: 27153112 PMCID: PMC4859540 DOI: 10.1371/journal.pone.0154982] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/23/2016] [Indexed: 01/04/2023] Open
Abstract
Oxygen consumption by isolated mitochondria is generally measured during state 4 respiration (no ATP production) or state 3 (maximal ATP production at high ADP availability). However, mitochondria in vivo do not function at either extreme. Here we used ADP recycling methodology to assess muscle mitochondrial function over intermediate clamped ADP concentrations. In so doing, we uncovered a previously unrecognized biphasic respiratory pattern wherein O2 flux on the complex II substrate, succinate, initially increased and peaked over low clamped ADP concentrations then decreased markedly at higher clamped concentrations. Mechanistic studies revealed no evidence that the observed changes in O2 flux were due to altered opening or function of the mitochondrial permeability transition pore or to changes in reactive oxygen. Based on metabolite and functional metabolic data, we propose a multifactorial mechanism that consists of coordinate changes that follow from reduced membrane potential (as the ADP concentration in increased). These changes include altered directional electron flow, altered NADH/NAD+ redox cycling, metabolite exit, and OAA inhibition of succinate dehydrogenase. In summary, we report a previously unrecognized pattern for complex II energized O2 flux. Moreover, our findings suggest that the ADP recycling approach might be more widely adapted for mitochondrial studies.
Collapse
Affiliation(s)
- Fan Bai
- Department of Biochemistry, University of Iowa, Iowa City, IA, 52242, United States of America
| | - Brian D. Fink
- Department of Internal Medicine / Endocrinology and Metabolism, University of Iowa and the Iowa City Veterans Affairs Medical Center, Iowa City, IA, 52242, United States of America
| | - Liping Yu
- Department of Biochemistry, University of Iowa, Iowa City, IA, 52242, United States of America
- NMR Core Facility, University of Iowa, Iowa City, IA, 52242, United States of America
| | - William I. Sivitz
- Department of Internal Medicine / Endocrinology and Metabolism, University of Iowa and the Iowa City Veterans Affairs Medical Center, Iowa City, IA, 52242, United States of America
- * E-mail:
| |
Collapse
|
30
|
Kaus A, Sareen D. ALS Patient Stem Cells for Unveiling Disease Signatures of Motoneuron Susceptibility: Perspectives on the Deadly Mitochondria, ER Stress and Calcium Triad. Front Cell Neurosci 2015; 9:448. [PMID: 26635528 PMCID: PMC4652136 DOI: 10.3389/fncel.2015.00448] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/02/2015] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a largely sporadic progressive neurodegenerative disease affecting upper and lower motoneurons (MNs) whose specific etiology is incompletely understood. Mutations in superoxide dismutase-1 (SOD1), TAR DNA-binding protein 43 (TARDBP/TDP-43) and C9orf72, have been identified in subsets of familial and sporadic patients. Key associated molecular and neuropathological features include ubiquitinated TDP-43 inclusions, stress granules, aggregated dipeptide proteins from mutant C9orf72 transcripts, altered mitochondrial ultrastructure, dysregulated calcium homeostasis, oxidative and endoplasmic reticulum (ER) stress, and an unfolded protein response (UPR). Such impairments have been documented in ALS animal models; however, whether these mechanisms are initiating factors or later consequential events leading to MN vulnerability in ALS patients is debatable. Human induced pluripotent stem cells (iPSCs) are a valuable tool that could resolve this “chicken or egg” causality dilemma. Relevant systems for probing pathophysiologically affected cells from large numbers of ALS patients and discovering phenotypic disease signatures of early MN susceptibility are described. Performing unbiased ‘OMICS and high-throughput screening in relevant neural cells from a cohort of ALS patient iPSCs, and rescuing mitochondrial and ER stress impairments, can identify targeted therapeutics for increasing MN longevity in ALS.
Collapse
Affiliation(s)
- Anjoscha Kaus
- Board of Governors-Regenerative Medicine Institute, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Biomedical Sciences, Cedars-Sinai Medical Center Los Angeles, CA, USA
| | - Dhruv Sareen
- Board of Governors-Regenerative Medicine Institute, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Biomedical Sciences, Cedars-Sinai Medical Center Los Angeles, CA, USA ; iPSC Core, The David and Janet Polak Stem Cell Laboratory, Cedars-Sinai Medical Center Los Angeles, CA, USA
| |
Collapse
|
31
|
Bell MT, Puskas F, Bennett DT, Cleveland JC, Herson PS, Mares JM, Meng X, Weyant MJ, Fullerton DA, Brett Reece T. Clinical indicators of paraplegia underplay universal spinal cord neuronal injury from transient aortic occlusion. Brain Res 2015; 1618:55-60. [DOI: 10.1016/j.brainres.2015.04.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 04/25/2015] [Accepted: 04/28/2015] [Indexed: 11/25/2022]
|
32
|
Metabolomics of Human Brain Aging and Age-Related Neurodegenerative Diseases. J Neuropathol Exp Neurol 2014; 73:640-57. [DOI: 10.1097/nen.0000000000000091] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
33
|
Fatty acids in energy metabolism of the central nervous system. BIOMED RESEARCH INTERNATIONAL 2014; 2014:472459. [PMID: 24883315 PMCID: PMC4026875 DOI: 10.1155/2014/472459] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 03/29/2014] [Accepted: 03/29/2014] [Indexed: 12/13/2022]
Abstract
In this review, we analyze the current hypotheses regarding energy metabolism in the neurons and astroglia. Recently, it was shown that up to 20% of the total brain's energy is provided by mitochondrial oxidation of fatty acids. However, the existing hypotheses consider glucose, or its derivative lactate, as the only main energy substrate for the brain. Astroglia metabolically supports the neurons by providing lactate as a substrate for neuronal mitochondria. In addition, a significant amount of neuromediators, glutamate and GABA, is transported into neurons and also serves as substrates for mitochondria. Thus, neuronal mitochondria may simultaneously oxidize several substrates. Astrocytes have to replenish the pool of neuromediators by synthesis de novo, which requires large amounts of energy. In this review, we made an attempt to reconcile β-oxidation of fatty acids by astrocytic mitochondria with the existing hypothesis on regulation of aerobic glycolysis. We suggest that, under condition of neuronal excitation, both metabolic pathways may exist simultaneously. We provide experimental evidence that isolated neuronal mitochondria may oxidize palmitoyl carnitine in the presence of other mitochondrial substrates. We also suggest that variations in the brain mitochondrial metabolic phenotype may be associated with different mtDNA haplogroups.
Collapse
|
34
|
Moser JM, Bigini P, Schmitt-John T. The wobbler mouse, an ALS animal model. Mol Genet Genomics 2013; 288:207-29. [PMID: 23539154 PMCID: PMC3664746 DOI: 10.1007/s00438-013-0741-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 03/12/2013] [Indexed: 12/11/2022]
Abstract
This review article is focused on the research progress made utilizing the wobbler mouse as animal model for human motor neuron diseases, especially the amyotrophic lateral sclerosis (ALS). The wobbler mouse develops progressive degeneration of upper and lower motor neurons and shows striking similarities to ALS. The cellular effects of the wobbler mutation, cellular transport defects, neurofilament aggregation, neuronal hyperexcitability and neuroinflammation closely resemble human ALS. Now, 57 years after the first report on the wobbler mouse we summarize the progress made in understanding the disease mechanism and testing various therapeutic approaches and discuss the relevance of these advances for human ALS. The identification of the causative mutation linking the wobbler mutation to a vesicle transport factor and the research focussed on the cellular basis and the therapeutic treatment of the wobbler motor neuron degeneration has shed new light on the molecular pathology of the disease and might contribute to the understanding the complexity of ALS.
Collapse
Affiliation(s)
- Jakob Maximilian Moser
- Molecular Biology and Genetics Department, Aarhus University, C. F. Møllers Alle 3, 8000 Aarhus C, Denmark
| | | | | |
Collapse
|
35
|
|
36
|
Abstract
Mutations in SOD1, causative for a subset of familial ALS cases, are associated with the formation of non-normal SOD1 conformers. Recent studies have defined this pool of SOD1 as misfolded and new antibodies have been developed to selectively detect misfolded SOD1 in vivo and in vitro. We will review these new tools and expand on the evidence demonstrating mitochondria as a common intersecting point for misfolded SOD1.
Collapse
Affiliation(s)
- Sarah Pickles
- Centre d'excellence en neuromique de l'Université de Montréal, Centre de recherche du CHUM (CRCHUM), Montréal, QC, Canada
| | | |
Collapse
|
37
|
Ferraiuolo L, Kirby J, Grierson AJ, Sendtner M, Shaw PJ. Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis. Nat Rev Neurol 2012; 7:616-30. [PMID: 22051914 DOI: 10.1038/nrneurol.2011.152] [Citation(s) in RCA: 459] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a genetically diverse disease. At least 15 ALS-associated gene loci have so far been identified, and the causative gene is known in approximately 30% of familial ALS cases. Less is known about the factors underlying the sporadic form of the disease. The molecular mechanisms of motor neuron degeneration are best understood in the subtype of disease caused by mutations in superoxide dismutase 1, with a current consensus that motor neuron injury is caused by a complex interplay between multiple pathogenic processes. A key recent finding is that mutated TAR DNA-binding protein 43 is a major constituent of the ubiquitinated protein inclusions in ALS, providing a possible link between the genetic mutation and the cellular pathology. New insights have also indicated the importance of dysregulated glial cell-motor neuron crosstalk, and have highlighted the vulnerability of the distal axonal compartment early in the disease course. In addition, recent studies have suggested that disordered RNA processing is likely to represent a major contributing factor to motor neuron disease. Ongoing research on the cellular pathways highlighted in this Review is predicted to open the door to new therapeutic interventions to slow disease progression in ALS.
Collapse
Affiliation(s)
- Laura Ferraiuolo
- Academic Neurology Unit, Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | | | | | | | | |
Collapse
|
38
|
Panov A, Kubalik N, Zinchenko N, Hemendinger R, Dikalov S, Bonkovsky HL. Respiration and ROS production in brain and spinal cord mitochondria of transgenic rats with mutant G93a Cu/Zn-superoxide dismutase gene. Neurobiol Dis 2011; 44:53-62. [PMID: 21745570 DOI: 10.1016/j.nbd.2011.06.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 05/30/2011] [Accepted: 06/09/2011] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Mitochondrial dysfunction is involved in the pathogenesis of motor neuron degeneration in the G93A mutant transgenic (tgmSOD1) animal model of ALS. However, it is unknown whether mitochondriopathy is a primary or secondary event. We isolated brain (BM) and spinal cord (SCM) mitochondria from 2 month old presymptomatic tgmSOD1 rats and studied respiration and generation of reactive oxygen species (ROS) using a new metabolic paradigm (Panov et al., Am. J. Physiol., Regul. Integr. Comp. Physiol., 2011). The yields of BM and SCM from tgmSOD1 rats were 27% and 58% lower than normal rats (WT). The rates of the State 3 and State 3U respiration of tgBM and tgSCM were normal with glutamate+pyruvate+malate as substrates but were inhibited with pyruvate+malate in tgBM and glutamate+malate in tgSCM. In tgSCM the State 4 respiration with all substrates was significantly (1.5-2 fold) increased as compared with WT-SCM. Western blot analysis showed that tgSCM had lower contents of complexes III (-60%) and IV (-35%), and the presence of mutated SOD1 protein in both tgBM and tgSCM. With glutamate+pyruvate+malate or succinate+glutamate+pyruvate+malate as substrates, tgBM and tgSCM generated 5-7 fold more ROS than normal mitochondria, and tgSCM generated two times more ROS than tgBM. We show that the major damaging ROS species in tgmSOD1 animals is H(2)O(2). It is known that mutated SOD1, damaged by H(2)O(2), associates with mitochondria, and we suggest that this further increases production of H(2)O(2). We also show that the total tissue calcium content remained normal in the brain but was diminished by 26% in the spinal cord of presymptomatic tgmSOD1 rats. CONCLUSION In tgSCM abnormally high rates of ROS generation, associated with reverse electron transport, result in accelerated mitochondriopathy, and the Ca(2+)-dependent excitotoxic death of motor neurons. Thus mitochondrial dysfunction is a key early element in pathogenesis of motor neuron degeneration in tgmSOD1 rats.
Collapse
Affiliation(s)
- Alexander Panov
- Carolinas Neuromuscular/ALS Research Laboratory, Cannon Research Center, Carolinas Medical Center, Charlotte, NC 28203, USA.
| | | | | | | | | | | |
Collapse
|