1
|
Leitner BP, Joseph P, Quast AF, Ramirez MA, Heerdt PM, Villalobos JG, Singh I. The metabolic and physiologic impairments underlying long COVID associated exercise intolerance. Pulm Circ 2024; 14:e70009. [PMID: 39544193 PMCID: PMC11560803 DOI: 10.1002/pul2.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/02/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024] Open
Abstract
Data from invasive CPET (iCPET) revealed long COVID patients have impaired systemic oxygen extraction (EO2), suggesting impaired mitochondrial ATP production. However, it remains uncertain whether the initial severity of SARS-CoV-2 infection has implications on EO2 and exercise capacity (VO2) nor has there been assessment of anerobic ATP generation in long COVID patients. iCPET was performed on 47 long COVID patients (i.e., full cohort; n = 8 with severe SARS-CoV-2 infection). In a subset of patients (i.e., metabolomic cohort; n = 26) metabolomics on venous and arterial blood samples during iCPET was performed. In the full cohort, long COVID patients exhibited reduced peak EO2 with reduced peak VO2 (90 ± 17% predicted) relative to cardiac output (118 ± 23% predicted). Peak VO2 [88% predicted (IQR 81% - 108%) vs. 70% predicted (IQR 64% - 89%); p = 0.02] and EO2 [0.59(IQR 0.53-0.62) vs. 0.53(IQR 0.50-0.48); p = 0.01) were lower in severe versus mild infection. In the metabolomic cohort, 12 metabolites were significantly consumed, and 41 metabolites were significantly released (p-values < 0.05). Quantitative metabolomics demonstrated significant increases in inosine and succinate arteriovenous gradients during exercise. Peak VO2 was significantly correlated with peak venous succinate (r = 0.68; p = 0.0008) and peak venous lactate (r = 0.49; p = 0.0004). Peak EO2 and consequently peak VO2 impact long COVID patients in a severity dependent manner. Exercise intolerance associated with long COVID is defined by impaired aerobic and anaerobic energy production. Peak venous succinate may serve as a potential biomarker in long COVID.
Collapse
Affiliation(s)
| | - Phillip Joseph
- Department of Medicine, Section of Pulmonary, Critical Care, and Sleep MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Andres Figueroa Quast
- Department of Medicine, Section of Pulmonary, Critical Care, and Sleep MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Maria Alejandra Ramirez
- Department of Medicine, Section of Pulmonary, Critical Care, and Sleep MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Paul M. Heerdt
- Department of AnesthesiologyYale School of MedicineNew HavenConnecticutUSA
| | - Jose G. Villalobos
- Department of Medicine, Section of Pulmonary, Critical Care, and Sleep MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Inderjit Singh
- Department of Medicine, Section of Pulmonary, Critical Care, and Sleep MedicineYale School of MedicineNew HavenConnecticutUSA
| |
Collapse
|
2
|
Zhang G, Mou Z, Wang H, Liu H. Comprehensive proteomic analysis of the main liver
and attached liver of <i>Glyptosternum maculatum</i> on the basis
of data-independent mass spectrometry acquisition. JOURNAL OF ANIMAL AND FEED SCIENCES 2022. [DOI: 10.22358/jafs/154070/2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
3
|
Yang L, Zhao Z, Luo D, Liang M, Zhang Q. Global Metabolomics of Fireflies (Coleoptera: Lampyridae) Explore Metabolic Adaptation to Fresh Water in Insects. INSECTS 2022; 13:823. [PMID: 36135524 PMCID: PMC9503472 DOI: 10.3390/insects13090823] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/01/2022] [Accepted: 09/03/2022] [Indexed: 06/16/2023]
Abstract
Aquatic insects are well-adapted to freshwater environments, but metabolic mechanisms of such adaptations, particularly to primary environmental factors (e.g., hypoxia, water pressure, dark light, and abundant microbes), are poorly known. Most firefly species (Coleoptera: Lampyridae) are terrestrial, but the larvae of a few species are aquatic. We generated 24 global metabolomic profiles of larvae and adults of Aquatica leii (freshwater) and Lychnuris praetexta (terrestrial) to identify freshwater adaptation-related metabolites (AARMs). We identified 110 differentially abundant metabolites (DAMs) in A. leii (adults vs. aquatic larvae) and 183 DAMs in L. praetexta (adults vs. terrestrial larvae). Furthermore, 100 DAMs specific to aquatic A. leii larvae were screened as AARMs via interspecific comparisons (A. leii vs. L. praetexta), which were primarily involved in antioxidant activity, immune response, energy production and metabolism, and chitin biosynthesis. They were assigned to six categories/superclasses (e.g., lipids and lipid-like molecules, organic acids and derivatives, and organoheterocyclic compound). Finally, ten metabolic pathways shared between KEGG terms specific to aquatic fireflies and enriched by AARMs were screened as aquatic adaptation-related pathways (AARPs). These AARPs were primarily involved in energy metabolism, xenobiotic biodegradation, protection of oxidative/immune damage, oxidative stress response, and sense function (e.g., glycine, serine and threonine metabolism, drug metabolism-cytochrome P450, and taste transduction), and certain aspects of morphology (e.g., steroid hormone biosynthesis). These results provide evidence suggesting that abundance changes in metabolomes contribute to freshwater adaptation of fireflies. The metabolites identified here may be vital targets for future work to determine the mechanism of freshwater adaptation in insects.
Collapse
Affiliation(s)
- Linyu Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Zishun Zhao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Dan Luo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
- YEN, Chuxiong People’s Hospital, Chuxiong 675000, China
| | - Mingzhong Liang
- Guangxi Key Laboratory of Marine Disaster in the Beibu Gulf, Ocean College, Beibu Gulf University, Qinzhou 535011, China
| | - Qilin Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
4
|
Semi-mechanistic Modeling of Hypoxanthine, Xanthine, and Uric Acid Metabolism in Asphyxiated Neonates. Clin Pharmacokinet 2022; 61:1545-1558. [PMID: 36040612 PMCID: PMC9652176 DOI: 10.1007/s40262-022-01164-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND OBJECTIVE Previously, we developed a pharmacokinetic-pharmacodynamic model of allopurinol, oxypurinol, and biomarkers, hypoxanthine, xanthine, and uric acid, in neonates with hypoxic-ischemic encephalopathy, in which high initial biomarker levels were observed suggesting an impact of hypoxia. However, the full pharmacodynamics could not be elucidated in our previous study. The current study included additional data from the ALBINO study (NCT03162653) placebo group, aiming to characterize the dynamics of hypoxanthine, xanthine, and uric acid in neonates with hypoxic-ischemic encephalopathy. METHODS Neonates from the ALBINO study who received allopurinol or placebo mannitol were included. An extended population pharmacokinetic-pharmacodynamic model was developed based on the mechanism of purine metabolism, where synthesis, salvage, and degradation via xanthine oxidoreductase pathways were described. The initial level of the biomarkers was a combination of endogenous turnover and high disease-related amounts. Model development was accomplished by nonlinear mixed-effects modeling (NONMEM®, version 7.5). RESULTS In total, 20 neonates treated with allopurinol and 17 neonates treated with mannitol were included in this analysis. Endogenous synthesis of the biomarkers reduced with 0.43% per hour because of precursor exhaustion. Hypoxanthine was readily salvaged or degraded to xanthine with rate constants of 0.5 1/h (95% confidence interval 0.33-0.77) and 0.2 1/h (95% confidence interval 0.09-0.31), respectively. A greater salvage was found in the allopurinol treatment group consistent with its mechanism of action. High hypoxia-induced initial levels of biomarkers were quantified, and were 1.2-fold to 2.9-fold higher in neonates with moderate-to-severe hypoxic-ischemic encephalopathy compared with those with mild hypoxic-ischemic encephalopathy. Half-maximal xanthine oxidoreductase inhibition was achieved with a combined allopurinol and oxypurinol concentration of 0.68 mg/L (95% confidence interval 0.48-0.92), suggesting full xanthine oxidoreductase inhibition during the period studied. CONCLUSIONS This extended pharmacokinetic-pharmacodynamic model provided an adequate description of the complex hypoxanthine, xanthine, and uric acid metabolism in neonates with hypoxic-ischemic encephalopathy, suggesting a positive allopurinol effect on these biomarkers. The impact of hypoxia on their dynamics was characterized, underlining higher hypoxia-related initial exposure with a more severe hypoxic-ischemic encephalopathy status.
Collapse
|
5
|
Genomewide analysis of sperm whale E2 ubiquitin conjugating enzyme genes. J Genet 2021. [DOI: 10.1007/s12041-021-01333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
6
|
A Study on Gentiana dahurica Fisch Ethanol Extract Alleviating Alcoholic Liver Disease in Mice: A Metabolomic Analysis of the Liver. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5569538. [PMID: 34257684 PMCID: PMC8260312 DOI: 10.1155/2021/5569538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 06/05/2021] [Accepted: 06/21/2021] [Indexed: 11/23/2022]
Abstract
We clarified the hepatoprotective effect of Gentiana dahurica Fisch ethanol extract (GDEE) in our previous study, and we further revealed the mechanism with the help of metabolomics technology in this study. The livers from Control group, Alcohol group, and Alcohol + GDEE group were analyzed by metabolomics. The metabolites in the liver were separated by ultra-high-performance liquid chromatography (UHPLC) and were tentatively identified using mass spectrometry (MS)/MS analysis. Differential metabolites were defined with VIP > 1 and P < 0.05. Principal component analysis (PCA) and orthogonal partial least square discriminant analysis (OPLS-DA) were applied to analyze differences among these groups. The results showed that the groups could be clearly distinguished by PCA and OPLS-DA analysis. Alcohol and GDEE could change the overall profile of liver metabolites. Alterations in liver tissues of ALD mice induced by alcohol were mainly involved in the dipeptides, purine and pyrimidine metabolism and glucose and lipid metabolism, which could be partly affected by GDEE. This study revealed that the mechanism of GDEE in alleviating ALD had the characteristics of multitarget and multipathway.
Collapse
|
7
|
Doğan HO, Şenol O, Bolat S, Yıldız ŞN, Büyüktuna SA, Sarıismailoğlu R, Doğan K, Hasbek M, Hekim SN. Understanding the pathophysiological changes via untargeted metabolomics in COVID-19 patients. J Med Virol 2020; 93:2340-2349. [PMID: 33300133 DOI: 10.1002/jmv.26716] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious respiratory disease caused by a new strain of the coronavirus. There is limited data on the pathogenesis and the cellular responses of COVID-19. In this study, we aimed to determine the variation of metabolites between healthy control and COVID-19 via the untargeted metabolomics method. Serum samples were obtained from 44 COVID-19 patients and 41 healthy controls. Untargeted metabolomics analyses were performed by the LC/Q-TOF/MS (liquid chromatography quadrupole time-of-flight mass spectrometry) method. Data acquisition, classification, and identification were achieved by the METLIN database and XCMS. Significant differences were determined between patients and healthy controls in terms of purine, glutamine, leukotriene D4 (LTD4), and glutathione metabolisms. Downregulations were determined in R-S lactoglutathione and glutamine. Upregulations were detected in hypoxanthine, inosine, and LTD4. Identified metabolites indicate roles for purine, glutamine, LTD4, and glutathione metabolisms in the pathogenesis of the COVID-19. The use of selective leukotriene D4 receptor antagonists, targeting purinergic signaling as a therapeutic approach and glutamine supplementation may decrease the severity and mortality of COVID-19.
Collapse
Affiliation(s)
- Halef O Doğan
- Department of Biochemistry, School of Medicine, University of Sivas Cumhuriyet, Sivas, Turkey
| | - Onur Şenol
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
| | - Serkan Bolat
- Department of Biochemistry, School of Medicine, University of Sivas Cumhuriyet, Sivas, Turkey
| | - Şeyma N Yıldız
- Department of Biochemistry, School of Medicine, University of Sivas Cumhuriyet, Sivas, Turkey
| | - Seyit A Büyüktuna
- Department of Infectious Diseases, School of Medicine, University of Sivas Cumhuriyet, Sivas, Turkey
| | | | - Kübra Doğan
- Department of Biochemistry, Sivas Numune Hospital, Sivas, Turkey
| | - Mürşit Hasbek
- Department of Microbiology, School of Medicine, University of Sivas Cumhuriyet, Sivas, Turkey
| | - Süleyman N Hekim
- Department of Biochemistry, School of Medicine, University of Biruni, İstanbul, Turkey
| |
Collapse
|
8
|
Effects of hypoxia and reoxygenation on intermediary metabolite homeostasis of marine bivalves Mytilus edulis and Crassostrea gigas. Comp Biochem Physiol A Mol Integr Physiol 2020; 242:110657. [DOI: 10.1016/j.cbpa.2020.110657] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 02/06/2023]
|
9
|
Allen KN, Vázquez-Medina JP. Natural Tolerance to Ischemia and Hypoxemia in Diving Mammals: A Review. Front Physiol 2019; 10:1199. [PMID: 31620019 PMCID: PMC6763568 DOI: 10.3389/fphys.2019.01199] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022] Open
Abstract
Reperfusion injury follows ischemia/reperfusion events occurring during myocardial infarction, stroke, embolism, and other peripheral vascular diseases. Decreased blood flow and reduced oxygen tension during ischemic episodes activate cellular pathways that upregulate pro-inflammatory signaling and promote oxidant generation. Reperfusion after ischemia recruits inflammatory cells to the vascular wall, further exacerbating oxidant production and ultimately resulting in cell death, tissue injury, and organ dysfunction. Diving mammals tolerate repetitive episodes of peripheral ischemia/reperfusion as part of the cardiovascular adjustments supporting long duration dives. These adjustments allow marine mammals to optimize the use of their body oxygen stores while diving but can result in selectively reduced perfusion to peripheral tissues. Remarkably, diving mammals show no apparent detrimental effects associated with these ischemia/reperfusion events. Here, we review the current knowledge regarding the strategies marine mammals use to suppress inflammation and cope with oxidant generation potentially derived from diving-induced ischemia/reperfusion.
Collapse
|
10
|
Apolipoprotein E4 and Insulin Resistance Interact to Impair Cognition and Alter the Epigenome and Metabolome. Sci Rep 2017; 7:43701. [PMID: 28272510 PMCID: PMC5341123 DOI: 10.1038/srep43701] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/30/2017] [Indexed: 02/07/2023] Open
Abstract
Apolipoprotein E4 (E4) and type 2 diabetes are major risk factors for cognitive decline and late onset Alzheimer's disease (AD). E4-associated phenotypes and insulin resistance (IR) share several features and appear to interact in driving cognitive dysfunction. However, shared mechanisms that could explain their overlapping pathophysiology have yet to be found. We hypothesized that, compared to E3 mice, E4 mice would be more susceptible to the harmful cognitive effects of high fat diet (HFD)-induced IR due to apoE isoform-specific differences in brain metabolism. While both E3 and E4 mice fed HFD displayed impairments in peripheral metabolism and cognition, deficits in hippocampal-dependent spatial learning and memory were exaggerated in E4 mice. Combining genome-wide measures of DNA hydroxymethylation with comprehensive untargeted metabolomics, we identified novel alterations in purine metabolism, glutamate metabolism, and the pentose phosphate pathway. Finally, in E4 mice, the metabolic and cognitive deficiencies caused by HFD were rescued by switching to a low fat diet for one month, suggesting a functional role was associated with reversal of the same metabolic pathways described above. These results suggest a susceptibility of E4 carriers to metabolic impairments brought on by IR, and may guide development of novel therapies for cognitive decline and dementia.
Collapse
|
11
|
Lindholm C, Altimiras J. Point-of-care devices for physiological measurements in field conditions. A smorgasbord of instruments and validation procedures. Comp Biochem Physiol A Mol Integr Physiol 2016; 202:99-111. [PMID: 27083239 DOI: 10.1016/j.cbpa.2016.04.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/30/2016] [Accepted: 04/09/2016] [Indexed: 01/05/2023]
Abstract
Point-of-care (POC) devices provide quick diagnostic results that increase the efficiency of patient care. Many POC devices are currently available to measure metabolites, blood gases, hormones, disease biomarkers or pathogens in samples as diverse as blood, urine, feces or exhaled breath. This diversity is potentially very useful for the comparative physiologist in field studies if proper validation studies are carried out to justify the accuracy of the devices in non-human species under different conditions. Our review presents an account of physiological parameters that can be monitored with POC devices and surveys the literature for suitable quantitative and statistical procedures for comparing POC measurements with reference "gold standard" procedures. We provide a set of quantitative tools and report on different correlation coefficients (Lin's Concordance Correlation Coefficient or the more widespread Pearson correlation coefficient), describe the graphical assessment of variation using Bland-Altman plots and discuss the difference between Model I and Model II regression procedures. We also report on three validation datasets for lactate, glucose and hemoglobin measurements in birds using the newly proposed procedures. We conclude the review with a haphazard account of future developments in the field, emphasizing the interest in lab-on-a-chip devices to carry out more complex experimental measurements than the ones currently available in POC devices.
Collapse
Affiliation(s)
- Caroline Lindholm
- Avian Behavioral Genomics and Physiology group, Division of Biology, Department of Physics, Chemistry and Biology (IFM), Linköping Univ., SE-58183 Linköping, Sweden
| | - Jordi Altimiras
- Avian Behavioral Genomics and Physiology group, Division of Biology, Department of Physics, Chemistry and Biology (IFM), Linköping Univ., SE-58183 Linköping, Sweden.
| |
Collapse
|