1
|
Huang K, Miao T, Dantas E, Han M, Hu Y, Wang K, Sanford J, Goncalves M, Perrimon N. Lipid metabolism of hepatocyte-like cells supports intestinal tumor growth by promoting tracheogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647255. [PMID: 40236168 PMCID: PMC11996582 DOI: 10.1101/2025.04.04.647255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Tumors require metabolic adaptations to support their rapid growth, but how they influence lipid metabolism in distant tissues remains poorly understood. Here, we uncover a novel mechanism by which gut tumors in adult flies reprogram lipid metabolism in distal hepatocyte-like cells, known as oenocytes, to promote tracheal development and tumor growth. We show that tumors secrete a PDGF/VEGF-like factor, Pvf1, that activates the TORC1-Hnf4 signaling pathway in oenocytes. This activation enhances the production of specific lipids, including very long-chain fatty acids and wax esters, that are required for tracheal growth surrounding the gut tumor. Importantly, reducing expression in oenocytes of either the transcription factor Hnf4 , or the elongase mElo that generates very long chain fatty acid suppresses tumor growth, tracheogenesis, and associated organ wasting/cachexia-like phenotypes, while extending lifespan. We further demonstrate that this regulatory pathway is conserved in mammals, as VEGF-A stimulates lipid metabolism gene expression in human hepatocytes, and lung tumor-bearing mice show increased hepatic expression of Hnf4 and the lipid elongation gene Elovl7 . Our findings reveal a previously unrecognized tumor-host interaction where tumors non-autonomously reprogram distal lipid metabolism to support their growth. This study not only identifies a novel non-autonomous role of the TORC1-Hnf4 axis in lipid-mediated tumor progression but also highlights potential targets for therapeutic intervention in cancer-associated metabolic disorders.
Collapse
|
2
|
Xiang F, Zhang Z, Xie J, Xiong S, Yang C, Liao D, Xia B, Lin L. Comprehensive review of the expanding roles of the carnitine pool in metabolic physiology: beyond fatty acid oxidation. J Transl Med 2025; 23:324. [PMID: 40087749 PMCID: PMC11907856 DOI: 10.1186/s12967-025-06341-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/01/2025] [Indexed: 03/17/2025] Open
Abstract
Traditionally, the carnitine pool is closely related to fatty acid metabolism. However, with increasing research, the pleiotropic effects of the carnitine pool have gradually emerged. The purpose of this review is to comprehensively investigate of the emerging understanding of the pleiotropic role of the carnitine pool, carnitine/acylcarnitines are not only auxiliaries or metabolites of fatty acid oxidation, but also play more complex and diverse roles, including energy metabolism, mitochondrial homeostasis, epigenetic regulation, regulation of inflammation and the immune system, tumor biology, signal transduction, and neuroprotection. This review provides an overview of the complex network of carnitine synthesis, transport, shuttle, and regulation, carnitine/acylcarnitines have the potential to be used as communication molecules, biomarkers and therapeutic targets for multiple diseases, with profound effects on intercellular communication, metabolic interactions between organs and overall metabolic health. The purpose of this review is to comprehensively summarize the multidimensional biological effects of the carnitine pool beyond its traditional role in fatty acid oxidation and to summarize the systemic effects mediated by carnitine/acylcarnitine to provide new perspectives for pharmacological research and treatment innovation and new strategies for the prevention and treatment of a variety of diseases.
Collapse
Affiliation(s)
- Feng Xiang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhimin Zhang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jingchen Xie
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Suhui Xiong
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chen Yang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Duanfang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Bohou Xia
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Limei Lin
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
3
|
Stoyanova S, Bogdanov MG. Rational Design, Synthesis, and In Vitro Activity of Heterocyclic Gamma-Butyrobetaines as Potential Carnitine Acetyltransferase Inhibitors. Molecules 2025; 30:735. [PMID: 39942839 PMCID: PMC11820905 DOI: 10.3390/molecules30030735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
This study investigates heterocyclic gamma-butyrobetaine (GBB) analogs as metabolic modulators through an integrated approach involving rational design, molecular docking, synthesis, and in vitro evaluation. The compounds synthesized demonstrated promising inhibitory potential toward carnitine acetyltransferase (CAT) and presumably other enzymes within the carnitine transferase family, with IC50 values ranging from 2.24 to 43.6 mM. Notably, some compounds demonstrated superior activity to the reference drug Meldonium (IC50 = 11.39 mM). A substantial outcome of the study that might serve as a foundation for future optimization and synthesis of more potent compounds was that a bulky, hydrophobic substituent at the gamma position enhances inhibitory activity, whereas esterification and increased polarity diminish it. The most effective compound was determined to be a reversible competitive inhibitor of CAT, with a Ki value of 3.5 mM comparable to Meldonium's Ki of 1.63 mM. These results suggest that heterocyclic GBB analogs present potential candidates for regulating metabolic processes and treating conditions including ischemic diseases, diabetes, and specific cancers.
Collapse
Affiliation(s)
| | - Milen G. Bogdanov
- Faculty of Chemistry and Pharmacy, Sofia University St. Kliment Ohridski, 1, Jammes Bourchier Blvd., 1164 Sofia, Bulgaria;
| |
Collapse
|
4
|
Reiss JD, Yang W, Chang AL, Long JZ, Marić I, Profit J, Sylvester KG, Stevenson DK, Aghaeepour N, Shaw GM. Transgenerational associations between newborn metabolic profiles and bronchopulmonary dysplasia in neonates born to mothers with an obese phenotype. Sci Rep 2025; 15:1144. [PMID: 39774255 PMCID: PMC11707363 DOI: 10.1038/s41598-025-85252-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
Maternal obesity increases risk for bronchopulmonary dysplasia (BPD) by up to 42%. Identifying metabolic features that may contribute to the association between maternal pre-pregnancy body mass index (BMI) and BPD is critical in defining the molecular relationship between these conditions. We investigated the association between maternal obesity and BPD using newborn screen metabolites as an explanatory variable. We hypothesized that elevated pre-pregnancy BMI compared to a normal BMI referent group, is associated with increased circulating short and long-chain acylcarnitines and subsequent development of BPD. This was a retrospective study with linkage of maternal pre-pregnancy BMI, with newborn screen metabolites obtained from the California Newborn Screening Program and further linked with neonatal outcomes. Results demonstrated elevated levels of phenylalanine and proline associated with an increased risk for BPD (OR 5.3, 95% CI 1.2-23.8 and OR 5.4, 95% CI 1.3-22.3) in the obesity group compared to the referent group. Short- and long-chain acylcarnitines demonstrated a mildly increased risk for BPD in neonates of mothers with severe obesity compared to controls. The findings suggest that specific metabolites may influence the molecular conditioning that increases susceptibility to BPD.
Collapse
Affiliation(s)
- Jonathan D Reiss
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA.
| | - Wei Yang
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
| | - Alan L Chang
- Stanford Department of Anesthesiology, Perioperative and Pain Medicine, 300 Pasteur Drive, Stanford, CA, USA
| | - Jonathan Z Long
- Department of Pathology and Stanford, Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University School of Medicine, Stanford, CA, USA
| | - Ivana Marić
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
| | - Jochen Profit
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
| | - Karl G Sylvester
- Stanford Department of Surgery, Division of Pediatric Surgery, 453 Quarry Rd, Palo Alto, CA, USA
| | - David K Stevenson
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
| | - Nima Aghaeepour
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
- Stanford Department of Anesthesiology, Perioperative and Pain Medicine, 300 Pasteur Drive, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Gary M Shaw
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
| |
Collapse
|
5
|
Lima V, Morais STB, Ferreira VG, Almeida MB, Silva MPB, de A. Lopes T, de Oliveira JM, Raimundo JRS, Furtado DZS, Fonseca FLA, Oliveira RV, Cardoso DR, Carrilho E, Assunção NA. Multiplatform Metabolomics: Enhancing the Severity Risk Prognosis of SARS-CoV-2 Infection. ACS OMEGA 2024; 9:45746-45758. [PMID: 39583673 PMCID: PMC11579725 DOI: 10.1021/acsomega.4c02557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024]
Abstract
Concerns about the SARS-CoV-2 outbreak (COVID-19) continue to persist even years later, with the emergence of new variants and the risk of disease severity. Common clinical symptoms, like cough, fever, and respiratory symptoms, characterize the noncritical patients, classifying them from mild to moderate. In a more severe and complex scenario, the virus infection can affect vital organs, resulting, for instance, in pneumonia and impaired kidney and heart function. However, it is well-known that subclinical symptoms at a metabolic level can be observed previously but require a proper diagnosis because viral replication on the host leaves a track with a different profile depending on the severity of the illness. Metabolomic profiles of mild, moderate, and severe COVID-19 patients were obtained by multiple platforms (LC-HRMS and MALDI-MS), increasing the chance to elucidate a prognosis for severity risk. A strong link was discovered between phenylalanine metabolism and increased COVID-19 severity symptoms, a pathway linked to cardiac and neurological consequences. Glycerophospholipids and sphingolipid metabolisms were also dysregulated linearly with the increasing symptom severity, which can be related to virus proliferation, immune system avoidance, and apoptosis escaping. Our data, endorsed by other literature, strengthens the notion that these pathways might play a vital role in a patient's prognosis.
Collapse
Affiliation(s)
- Vinicius
S. Lima
- Programa
de Pós-Graduação em Medicina Translacional, Departamento
de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | - Sinara T. B. Morais
- Instituto
de Química de São Carlos, Universidade de São Paulo, São Carlos 13566-590, Brazil
| | - Vinicius G. Ferreira
- Instituto
de Química de São Carlos, Universidade de São Paulo, São Carlos 13566-590, Brazil
- Instituto
Nacional de Ciência e Tecnologia de Bioanalítica, INCTBio, Campinas 13083-861, Brazil
| | - Mariana B. Almeida
- Instituto
de Química de São Carlos, Universidade de São Paulo, São Carlos 13566-590, Brazil
- Instituto
Nacional de Ciência e Tecnologia de Bioanalítica, INCTBio, Campinas 13083-861, Brazil
| | - Manuel Pedro Barros Silva
- Programa
de Pós-Graduação em Medicina Translacional, Departamento
de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | - Thais de A. Lopes
- Departamento
de Química, Universidade Federal
de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Juliana M. de Oliveira
- Departamento
de Química, Universidade Federal
de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | | | - Danielle Z. S. Furtado
- Programa
de Pós-Graduação em Medicina Translacional, Departamento
de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | - Fernando L. A. Fonseca
- Faculdade
de Medicina do ABC, Santo André, São Paulo 09060-870, Brazil
- Departamento
de Química, Universidade Federal
de São Paulo, São
Paulo 05508-070, Brazil
| | - Regina V. Oliveira
- Departamento
de Química, Universidade Federal
de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Daniel R. Cardoso
- Instituto
de Química de São Carlos, Universidade de São Paulo, São Carlos 13566-590, Brazil
| | - Emanuel Carrilho
- Instituto
de Química de São Carlos, Universidade de São Paulo, São Carlos 13566-590, Brazil
- Instituto
Nacional de Ciência e Tecnologia de Bioanalítica, INCTBio, Campinas 13083-861, Brazil
| | - Nilson A. Assunção
- Programa
de Pós-Graduação em Medicina Translacional, Departamento
de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
- Departamento
de Química, Universidade Federal
de São Paulo, São
Paulo 05508-070, Brazil
| |
Collapse
|
6
|
Şensoy E. Melatonin prevents histopathologies stem from cadmium chloride in pregnant mice lungs. J Mol Histol 2024; 55:955-965. [PMID: 39198364 DOI: 10.1007/s10735-024-10243-z] [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: 05/28/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024]
Abstract
Heavy metals may cause structural and functional changes in organs. Cadmium, taken into the body through oral and respiratory routes, can lead to lesions. Cadmium may lead to lesions by accumulating in organs. The lungs are significantly affected by cadmium. Melatonin, an antioxidant hormone with therapeutic effects, is secreted by the pineal gland. The aim of the study is to treat cadmium-induced lesions in the lungs of pregnant mice with Melatonin. Four groups were created with 24 pregnant mice, named Control, Cadmium Chloride, Melatonin, and Melatonin + Cadmium Chloride groups (n: 6) Cadmium Chloride (2 mg/kg/bw) and Melatonin (3 mg/kg/bw) were given orally through gavage during pregnancy (21 days) After routine histological procedures, the lung tissues were stained with Hematoxylin-Eosin and evaluated under a light and electron microscope. ANOVA tests were applied for one-way analysis of variance, and LSD tests were applied for pairwise comparisons (p < 0.05) The average lung weight decreased in the Cadmium Chloride group (p: 0.03) The average lung weight in the Cadmium Chloride + Melatonin group was found to be close to the control group (p: 0.06) Cadmium Chloride caused thickening of the lung alveolar wall, inflammatory cell infiltration, and fibrin deposition. Because the lesions were not observed in the Melatonin group, lesions may be prevented by melatonin. Additional studies may be useful to determine the protective effect of Melatonin at different doses of Cadmium Chloride.
Collapse
Affiliation(s)
- Erhan Şensoy
- Department of Midwifery, Faculty of Health Sciences, KaramanogluMehmetbey University, Karaman, Turkey.
| |
Collapse
|
7
|
Chung KP, Cheng CN, Chen YJ, Hsu CL, Huang YL, Hsieh MS, Kuo HC, Lin YT, Juan YH, Nakahira K, Chen YF, Liu WL, Ruan SY, Chien JY, Plataki M, Cloonan SM, Carmeliet P, Choi AMK, Kuo CH, Yu CJ. Alveolar epithelial cells mitigate neutrophilic inflammation in lung injury through regulating mitochondrial fatty acid oxidation. Nat Commun 2024; 15:7241. [PMID: 39174557 PMCID: PMC11341863 DOI: 10.1038/s41467-024-51683-1] [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: 10/01/2023] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Type 2 alveolar epithelial (AT2) cells of the lung are fundamental in regulating alveolar inflammation in response to injury. Impaired mitochondrial long-chain fatty acid β-oxidation (mtLCFAO) in AT2 cells is assumed to aggravate alveolar inflammation in acute lung injury (ALI), yet the importance of mtLCFAO to AT2 cell function needs to be defined. Here we show that expression of carnitine palmitoyltransferase 1a (CPT1a), a mtLCFAO rate limiting enzyme, in AT2 cells is significantly decreased in acute respiratory distress syndrome (ARDS). In mice, Cpt1a deletion in AT2 cells impairs mtLCFAO without reducing ATP production and alters surfactant phospholipid abundance in the alveoli. Impairing mtLCFAO in AT2 cells via deleting either Cpt1a or Acadl (acyl-CoA dehydrogenase long chain) restricts alveolar inflammation in ALI by hindering the production of the neutrophilic chemokine CXCL2 from AT2 cells. This study thus highlights mtLCFAO as immunometabolism to injury in AT2 cells and suggests impaired mtLCFAO in AT2 cells as an anti-inflammatory response in ARDS.
Collapse
Grants
- K08 HL157728 NHLBI NIH HHS
- 109-O04, 110-O07, 110-S4872, 111-S0075, 113-S0079 National Taiwan University Hospital (NTUH)
- NTUCDP-112L7745, NTUCDP-112L7746, 110T099, NTU-NFG-110L7422 National Taiwan University (NTU)
- National Science and Technology Council (Taiwan) (MOST-108-2628-B-002-017 [K.P.C.], MOST-109-2628-B-002-044 [K.P.C.], MOST-110-2628-B-002-029 [K.P.C.], MOST-110-2628-B-002-045-MY3 [K.P.C.], MOST-111-2628-B-002-030-MY3 [K.P.C.])
- National Science and Technology Council (Taiwan), MOST 107-2314-B-002-235-MY3
- National Science and Technology Council (Taiwan), MOST 110-2314-B-002-262
- National Taiwan University School of Pharmacy Endowment Fund in support of the Platform for Clinical Mass Spectrometry and NMR Structure Elucidation
- Research funding provided by Mr. Barry Lam, the chairman of Quanta Computer Inc
Collapse
Affiliation(s)
- Kuei-Pin Chung
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chih-Ning Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Jung Chen
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Lin Huang
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Chun Kuo
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Ting Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Hsiu Juan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kiichi Nakahira
- Department of Pharmacology, Nara Medical University, Kashihara, Nara, Japan
| | - Yen-Fu Chen
- Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Wei-Lun Liu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
- Department of Critical Care Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei, Taiwan
| | - Sheng-Yuan Ruan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jung-Yien Chien
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chong-Jen Yu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan.
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
8
|
Narayanan B, Xia C, McAndrew R, Shen AL, Kim JJP. Structural basis for expanded substrate specificities of human long chain acyl-CoA dehydrogenase and related acyl-CoA dehydrogenases. Sci Rep 2024; 14:12976. [PMID: 38839792 PMCID: PMC11153573 DOI: 10.1038/s41598-024-63027-6] [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: 02/23/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024] Open
Abstract
Crystal structures of human long-chain acyl-CoA dehydrogenase (LCAD) and the catalytically inactive Glu291Gln mutant, have been determined. These structures suggest that LCAD harbors functions beyond its historically defined role in mitochondrial β-oxidation of long and medium-chain fatty acids. LCAD is a homotetramer containing one FAD per 43 kDa subunit with Glu291 as the catalytic base. The substrate binding cavity of LCAD reveals key differences which makes it specific for longer and branched chain substrates. The presence of Pro132 near the start of the E helix leads to helix unwinding that, together with adjacent smaller residues, permits binding of bulky substrates such as 3α, 7α, l2α-trihydroxy-5β-cholestan-26-oyl-CoA. This structural element is also utilized by ACAD11, a eucaryotic ACAD of unknown function, as well as bacterial ACADs known to metabolize sterol substrates. Sequence comparison suggests that ACAD10, another ACAD of unknown function, may also share this substrate specificity. These results suggest that LCAD, ACAD10, ACAD11 constitute a distinct class of eucaryotic acyl CoA dehydrogenases.
Collapse
Affiliation(s)
- Beena Narayanan
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Chuanwu Xia
- Department of Chemistry and Biochemistry, College of Arts and Sciences, University of North Florida, Jacksonville, FL, 32224, USA
| | - Ryan McAndrew
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, 94740, USA
| | - Anna L Shen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jung-Ja P Kim
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
9
|
Babcock SJ, Houten SM, Gillingham MB. A review of fatty acid oxidation disorder mouse models. Mol Genet Metab 2024; 142:108351. [PMID: 38430613 PMCID: PMC11073919 DOI: 10.1016/j.ymgme.2024.108351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2024]
Abstract
Fatty acid oxidation disorders (FAODs) are a family of rare, genetic disorders that affect any part of the fatty acid oxidation pathway. Patients present with severe phenotypes, such as hypoketotic hypoglycemia, cardiomyopathy, and rhabdomyolysis, and currently manage these symptoms by the avoidance of fasting and maintaining a low-fat, high-carbohydrate diet. Because knowledge about FAODs is limited due to the small number of patients, rodent models have been crucial in learning more about these disorders, particularly in studying the molecular mechanisms involved in different phenotypes and in evaluating treatments for patients. The purpose of this review is to present the different FAOD mouse models and highlight the benefits and limitations of using these models. Specifically, we discuss the phenotypes of the available FAOD mouse models, the potential molecular causes of prominent FAOD phenotypes that have been studied using FAOD mouse models, and how FAOD mouse models have been used to evaluate treatments for patients.
Collapse
Affiliation(s)
- Shannon J Babcock
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA.
| | - Sander M Houten
- Deparment of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Melanie B Gillingham
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
10
|
Narayanan B, Xia C, McAndrew R, Shen AL, Kim JJP. Structural Basis for Expanded Substrate Speci ficities of Human Long Chain Acyl-CoA Dehydrogenase and Related Acyl- CoA Dehydrogenases. RESEARCH SQUARE 2024:rs.3.rs-3980524. [PMID: 38464032 PMCID: PMC10925408 DOI: 10.21203/rs.3.rs-3980524/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Crystal structures of human long-chain acyl-CoA dehydrogenase (LCAD) and the E291Q mutant, have been determined. These structures suggest that LCAD harbors functions beyond its historically defined role in mitochondrial β-oxidation of long and medium-chain fatty acids. LCAD is a homotetramer containing one FAD per 43kDa subunit with Glu291 as the catalytic base. The substrate binding cavity of LCAD reveals key differences which makes it specific for longer and branched chain substrates. The presence of Pro132 near the start of the E helix leads to helix unwinding that, together with adjacent smaller residues, permits binding of bulky substrates such as 3α, 7α, l2α-trihydroxy-5β-cholestan-26-oyl-CoA. This structural element is also utilized by ACAD11, a eucaryotic ACAD of unknown function, as well as bacterial ACADs known to metabolize sterol substrates. Sequence comparison suggests that ACAD10, another ACAD of unknown function, may also share this substrate specificity. These results suggest that LCAD, ACAD10, ACAD11 constitute a distinct class of eucaryotic acyl CoA dehydrogenases.
Collapse
|
11
|
El-Derany MO, Hanna DMF, Youshia J, Elmowafy E, Farag MA, Azab SS. Metabolomics-directed nanotechnology in viral diseases management: COVID-19 a case study. Pharmacol Rep 2023; 75:1045-1065. [PMID: 37587394 PMCID: PMC10539420 DOI: 10.1007/s43440-023-00517-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/18/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently regarded as the twenty-first century's plague accounting for coronavirus disease 2019 (COVID-19). Besides its reported symptoms affecting the respiratory tract, it was found to alter several metabolic pathways inside the body. Nanoparticles proved to combat viral infections including COVID-19 to demonstrate great success in developing vaccines based on mRNA technology. However, various types of nanoparticles can affect the host metabolome. Considering the increasing proportion of nano-based vaccines, this review compiles and analyses how COVID-19 and nanoparticles affect lipids, amino acids, and carbohydrates metabolism. A search was conducted on PubMed, ScienceDirect, Web of Science for available information on the interrelationship between metabolomics and immunity in the context of SARS-CoV-2 infection and the effect of nanoparticles on metabolite levels. It was clear that SARS-CoV-2 disrupted several pathways to ensure a sufficient supply of its building blocks to facilitate its replication. Such information can help in developing treatment strategies against viral infections and COVID-19 based on interventions that overcome these metabolic changes. Furthermore, it showed that even drug-free nanoparticles can exert an influence on biological systems as evidenced by metabolomics.
Collapse
Affiliation(s)
- Marwa O El-Derany
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Diana M F Hanna
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| | - John Youshia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Enas Elmowafy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr El-Aini St., P.B. 11562, Cairo, Egypt
| | - Samar S Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt.
| |
Collapse
|
12
|
Vianey-Saban C, Guffon N, Fouilhoux A, Acquaviva C. Fifty years of research on mitochondrial fatty acid oxidation disorders: The remaining challenges. J Inherit Metab Dis 2023; 46:848-873. [PMID: 37530674 DOI: 10.1002/jimd.12664] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023]
Abstract
Since the identification of the first disorder of mitochondrial fatty acid oxidation defects (FAOD) in 1973, more than 20 defects have been identified. Although there are some differences, most FAOD have similar clinical signs, which are mainly due to energy depletion and toxicity of accumulated metabolites. However, some of them have an unusual clinical phenotype or specific clinical signs. This manuscript focuses on what we have learnt so far on the pathophysiology of these disorders, which present with clinical signs that are not typical of categorical FAOD. It also highlights that some disorders have not yet been identified and tries to make assumptions to explain why. It also deals with new treatments under consideration in FAOD, including triheptanoin and similar anaplerotic substrates, ketone body treatments, RNA and gene therapy approaches. Finally, it suggests challenges for the diagnosis of FAOD in the coming years, both for symptomatic patients and for those diagnosed through newborn screening. The ultimate goal would be to identify all the patients born with FAOD and ensure for them the best possible quality of life.
Collapse
Affiliation(s)
- Christine Vianey-Saban
- Biochemical and Molecular Biology Laboratory, Metabolic Inborn Errors of Metabolism Unit, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Nathalie Guffon
- National Reference Centre for Hereditary Metabolic Diseases, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Alain Fouilhoux
- National Reference Centre for Hereditary Metabolic Diseases, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Cécile Acquaviva
- Biochemical and Molecular Biology Laboratory, Metabolic Inborn Errors of Metabolism Unit, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| |
Collapse
|
13
|
den Hartog I, Karu N, Zwep LB, Voorn GP, van de Garde EM, Hankemeier T, van Hasselt JC. Differential metabolic host response to pathogens associated with community-acquired pneumonia. Metabol Open 2023; 18:100239. [PMID: 37025095 PMCID: PMC10070890 DOI: 10.1016/j.metop.2023.100239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Background Metabolic changes induced by the host immune response to pathogens found in patients with community-acquired pneumonia (CAP) may provide insight into its pathogenesis. In this study, we characterized differences in the host metabolic response to common CAP-associated pathogens. Method Targeted metabolomic profiling was performed on serum samples obtained from hospitalized CAP patients (n = 119) at admission. We quantified 347 unique metabolites across multiple biochemical classes, including amines, acylcarnitines, and signaling lipids. We evaluated if unique associations between metabolite levels and specific CAP-associated pathogens could be identified. Results Several acylcarnitines were found to be elevated in C. burnetii and herpes simplex virus and lowered in M. pneumoniae as compared to other pathogens. Phenylalanine and kynurenine were found elevated in L. pneumophila as compared to other pathogens. S-methylcysteine was elevated in patients with M. pneumoniae, and these patients also showed lowered cortisol levels in comparison to almost all other pathogens. For the herpes simplex virus, we observed a unique elevation of eicosanoids and several amines. Many lysophosphatidylcholines showed an altered profile in C. burnetii versus S. pneumoniae, L. pneumophila, and respiratory syncytial virus. Finally, phosphatidylcholines were negatively affected by the influenza virus in comparison to S. pneumoniae. Conclusions In this exploratory analysis, metabolites from different biochemical classes were found to be altered in serum samples from patients with different CAP-associated pathogens, which may be used for hypothesis generation in studies on differences in pathogen host response and pathogenesis of CAP.
Collapse
Affiliation(s)
- Ilona den Hartog
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Naama Karu
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Laura B. Zwep
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - G. Paul Voorn
- Department of Medical Microbiology and Immunology, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Ewoudt M.W. van de Garde
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - J.G. Coen van Hasselt
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
- Corresponding author.
| |
Collapse
|
14
|
Suber TL, Wendell SG, Mullett SJ, Zuchelkowski B, Bain W, Kitsios GD, McVerry BJ, Ray P, Ray A, Mallampalli RK, Zhang Y, Shah F, Nouraie SM, Lee JS. Serum metabolomic signatures of fatty acid oxidation defects differentiate host-response subphenotypes of acute respiratory distress syndrome. Respir Res 2023; 24:136. [PMID: 37210531 PMCID: PMC10199668 DOI: 10.1186/s12931-023-02447-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 05/09/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Fatty acid oxidation (FAO) defects have been implicated in experimental models of acute lung injury and associated with poor outcomes in critical illness. In this study, we examined acylcarnitine profiles and 3-methylhistidine as markers of FAO defects and skeletal muscle catabolism, respectively, in patients with acute respiratory failure. We determined whether these metabolites were associated with host-response ARDS subphenotypes, inflammatory biomarkers, and clinical outcomes in acute respiratory failure. METHODS In a nested case-control cohort study, we performed targeted analysis of serum metabolites of patients intubated for airway protection (airway controls), Class 1 (hypoinflammatory), and Class 2 (hyperinflammatory) ARDS patients (N = 50 per group) during early initiation of mechanical ventilation. Relative amounts were quantified by liquid chromatography high resolution mass spectrometry using isotope-labeled standards and analyzed with plasma biomarkers and clinical data. RESULTS Of the acylcarnitines analyzed, octanoylcarnitine levels were twofold increased in Class 2 ARDS relative to Class 1 ARDS or airway controls (P = 0.0004 and < 0.0001, respectively) and was positively associated with Class 2 by quantile g-computation analysis (P = 0.004). In addition, acetylcarnitine and 3-methylhistidine were increased in Class 2 relative to Class 1 and positively correlated with inflammatory biomarkers. In all patients within the study with acute respiratory failure, increased 3-methylhistidine was observed in non-survivors at 30 days (P = 0.0018), while octanoylcarnitine was increased in patients requiring vasopressor support but not in non-survivors (P = 0.0001 and P = 0.28, respectively). CONCLUSIONS This study demonstrates that increased levels of acetylcarnitine, octanoylcarnitine, and 3-methylhistidine distinguish Class 2 from Class 1 ARDS patients and airway controls. Octanoylcarnitine and 3-methylhistidine were associated with poor outcomes in patients with acute respiratory failure across the cohort independent of etiology or host-response subphenotype. These findings suggest a role for serum metabolites as biomarkers in ARDS and poor outcomes in critically ill patients early in the clinical course.
Collapse
Affiliation(s)
- Tomeka L Suber
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA.
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Stacy G Wendell
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steven J Mullett
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Benjamin Zuchelkowski
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - William Bain
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Georgios D Kitsios
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bryan J McVerry
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Prabir Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anuradha Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rama K Mallampalli
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - Faraaz Shah
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Seyed Mehdi Nouraie
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - Janet S Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University at St. Louis, St. Louis, MO, USA
| |
Collapse
|
15
|
Smith MR, Hu X, Jarrell ZR, He X, Orr M, Fernandes J, Chandler JD, Walker DI, Esper A, Marts L, Neujahr DC, Jones DP, Go YM. Study on the Relationship between Selenium and Cadmium in Diseased Human Lungs. ADVANCES IN REDOX RESEARCH 2023; 7. [PMID: 37034445 PMCID: PMC10078579 DOI: 10.1016/j.arres.2023.100065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Cadmium (Cd) is a toxic environmental metal that interacts with selenium (Se) and contributes to many lung diseases. Humans have widespread exposures to Cd through diet and cigarette smoking, and studies in rodent models show that Se can protect against Cd toxicities. We sought to identify whether an antagonistic relationship existed between Se and Cd burdens and determine whether this relationship may associate with metabolic variation within human lungs. We performed metabolomics of 31 human lungs, including 25 with end-stage lung disease due to idiopathic pulmonary fibrosis, cystic fibrosis, chronic obstructive lung disease (COPD)/emphysema and other causes, and 6 non-diseased lungs. Results showed pathway associations with Cd including amino acid, lipid and energy-related pathways. Metabolic pathways varying with Se had considerable overlap with these pathways. Hierarchical cluster analysis (HCA) of individuals according to metabolites associated with Cd showed partial separation of disease types, with COPD/emphysema in the cluster with highest Cd, and non-diseased lungs in the cluster with the lowest Cd. When compared to HCA of metabolites associated with Se, the results showed that the cluster containing COPD/emphysema had the lowest Se, and the non-diseased lungs had the highest Se. A greater number of pathway associations occurred for Cd to Se ratio than either Cd or Se alone, indicating that metabolic patterns were more dependent on Cd to Se ratio than on either alone. Network analysis of interactions of Cd and Se showed network centrality was associated with pathways linked to polyunsaturated fatty acids involved in inflammatory signaling. Overall, the data show that metabolic pathway responses in human lung vary with Cd and Se in a pattern suggesting that Se is antagonistic to Cd toxicity in humans.
Collapse
Affiliation(s)
- Matthew Ryan Smith
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
- Atlanta Department of Veterans Affairs Medical Center, Decatur, GA, USA
| | - Xin Hu
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
| | - Zachery R Jarrell
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
| | - Xiaojia He
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
| | - Michael Orr
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
| | - Jolyn Fernandes
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Joshua D. Chandler
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
- Department of Pediatrics, School of Medicine at Emory University, Atlanta, GA, USA
| | - Douglas I. Walker
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Annette Esper
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
| | - Lucian Marts
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
| | - David C. Neujahr
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
| | - Dean P. Jones
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
- Corresponding authors at: Whitehead Biomedical Research Building, 615 Michael St, Room 225, Atlanta, GA, 30322, USA. (D.P. Jones), (Y.-M. Go)
| | - Young-Mi Go
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine at Emory University, Atlanta, GA, USA
- Corresponding authors at: Whitehead Biomedical Research Building, 615 Michael St, Room 225, Atlanta, GA, 30322, USA. (D.P. Jones), (Y.-M. Go)
| |
Collapse
|
16
|
Moura AV, de Oliveira DC, Silva AAR, da Rosa JR, Garcia PHD, Sanches PHG, Garza KY, Mendes FMM, Lambert M, Gutierrez JM, Granado NM, dos Santos AC, de Lima IL, Negrini LDDO, Antonio MA, Eberlin MN, Eberlin LS, Porcari AM. Urine Metabolites Enable Fast Detection of COVID-19 Using Mass Spectrometry. Metabolites 2022; 12:1056. [PMID: 36355139 PMCID: PMC9697918 DOI: 10.3390/metabo12111056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 08/27/2023] Open
Abstract
The COVID-19 pandemic boosted the development of diagnostic tests to meet patient needs and provide accurate, sensitive, and fast disease detection. Despite rapid advancements, limitations related to turnaround time, varying performance metrics due to different sampling sites, illness duration, co-infections, and the need for particular reagents still exist. As an alternative diagnostic test, we present urine analysis through flow-injection-tandem mass spectrometry (FIA-MS/MS) as a powerful approach for COVID-19 diagnosis, targeting the detection of amino acids and acylcarnitines. We adapted a method that is widely used for newborn screening tests on dried blood for urine samples in order to detect metabolites related to COVID-19 infection. We analyzed samples from 246 volunteers with diagnostic confirmation via PCR. Urine samples were self-collected, diluted, and analyzed with a run time of 4 min. A Lasso statistical classifier was built using 75/25% data for training/validation sets and achieved high diagnostic performances: 97/90% sensitivity, 95/100% specificity, and 95/97.2% accuracy. Additionally, we predicted on two withheld sets composed of suspected hospitalized/symptomatic COVID-19-PCR negative patients and patients out of the optimal time-frame collection for PCR diagnosis, with promising results. Altogether, we show that the benchmarked FIA-MS/MS method is promising for COVID-19 screening and diagnosis, and is also potentially useful after the peak viral load has passed.
Collapse
Affiliation(s)
- Alexandre Varao Moura
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Danilo Cardoso de Oliveira
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Alex Ap. R. Silva
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Jonas Ribeiro da Rosa
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Pedro Henrique Dias Garcia
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Pedro Henrique Godoy Sanches
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Kyana Y. Garza
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA
| | - Flavio Marcio Macedo Mendes
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Mayara Lambert
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Junier Marrero Gutierrez
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Nicole Marino Granado
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Alicia Camacho dos Santos
- Department of Material Engineering and Nanotechnology, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil
| | - Iasmim Lopes de Lima
- Department of Material Engineering and Nanotechnology, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil
| | | | - Marcia Aparecida Antonio
- Integrated Unit of Pharmacology and Gastroenterology, UNIFAG, Bragança Paulista 12916-900, SP, Brazil
| | - Marcos N. Eberlin
- Department of Material Engineering and Nanotechnology, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil
| | - Livia S. Eberlin
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andreia M. Porcari
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| |
Collapse
|
17
|
Kelly RS, Stewart ID, Bayne H, Kachroo P, Spiro A, Vokonas P, Sparrow D, Weiss ST, Knihtilä HM, Litonjua AA, Wareham NJ, Langenberg C, Lasky-Su JA. Metabolomic differences in lung function metrics: evidence from two cohorts. Thorax 2022; 77:919-928. [PMID: 34650005 PMCID: PMC9008068 DOI: 10.1136/thoraxjnl-2020-216639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 09/18/2021] [Indexed: 12/20/2022]
Abstract
RATIONALE The biochemical mechanisms underlying lung function are incompletely understood. OBJECTIVES To identify and validate the plasma metabolome of lung function using two independent adult cohorts: discovery-the European Prospective Investigation into Cancer-Norfolk (EPIC-Norfolk, n=10 460) and validation-the VA Normative Aging Study (NAS) metabolomic cohort (n=437). METHODS We ran linear regression models for 693 metabolites to identify associations with forced expiratory volume in one second (FEV1) and the ratio of FEV1 to forced vital capacity (FEV1/FVC), in EPIC-Norfolk then validated significant findings in NAS. Significance in EPIC-Norfolk was denoted using an effective number of tests threshold of 95%; a metabolite was considered validated in NAS if the direction of effect was consistent and p<0.05. MEASUREMENTS AND MAIN RESULTS Of 156 metabolites that associated with FEV1 in EPIC-Norfolk after adjustment for age, sex, body mass index, height, smoking and asthma status, 34 (21.8%) validated in NAS, including several metabolites involved in oxidative stress. When restricting the discovery sample to men only, a similar percentage, 18 of 79 significant metabolites (22.8%) were validated. A smaller number of metabolites were validated for FEV1/FVC, 6 of 65 (9.2%) when including all EPIC-Norfolk as the discovery population, and 2 of 34 (5.9%) when restricting to men. These metabolites were characterised by involvement in respiratory track secretants. Interestingly, no metabolites were validated for both FEV1 and FEV1/FVC. CONCLUSIONS The validation of metabolites associated with respiratory function can help to better understand mechanisms of lung health and may assist the development of biomarkers.
Collapse
Affiliation(s)
- Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Haley Bayne
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Priyadarshini Kachroo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Avron Spiro
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), 150 South Huntington Avenue, Boston, MA 02130, USA, VA Boston Healthcare System, Boston, MA 02130, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Pantel Vokonas
- VA Normative Aging Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - David Sparrow
- VA Normative Aging Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Hanna M Knihtilä
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Ye C, Wu J, Reiss JD, Sinclair TJ, Stevenson DK, Shaw GM, Chace DH, Clark RH, Prince LS, Ling XB, Sylvester KG. Progressive Metabolic Abnormalities Associated with the Development of Neonatal Bronchopulmonary Dysplasia. Nutrients 2022; 14:nu14173547. [PMID: 36079804 PMCID: PMC9459725 DOI: 10.3390/nu14173547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Objective: To assess the longitudinal metabolic patterns during the evolution of bronchopulmonary dysplasia (BPD) development. Methods: A case-control dataset of preterm infants (<32-week gestation) was obtained from a multicenter database, including 355 BPD cases and 395 controls. A total of 72 amino acid (AA) and acylcarnitine (AC) variables, along with infants’ calorie intake and growth outcomes, were measured on day of life 1, 7, 28, and 42. Logistic regression, clustering methods, and random forest statistical modeling were utilized to identify metabolic variables significantly associated with BPD development and to investigate their longitudinal patterns that are associated with BPD development. Results: A panel of 27 metabolic variables were observed to be longitudinally associated with BPD development. The involved metabolites increased from 1 predominant different AC by day 7 to 19 associated AA and AC compounds by day 28 and 16 metabolic features by day 42. Citrulline, alanine, glutamate, tyrosine, propionylcarnitine, free carnitine, acetylcarnitine, hydroxybutyrylcarnitine, and most median-chain ACs (C5:C10) were the most associated metabolites down-regulated in BPD babies over the early days of life, whereas phenylalanine, methionine, and hydroxypalmitoylcarnitine were observed to be up-regulated in BPD babies. Most calorie intake and growth outcomes revealed similar longitudinal patterns between BPD cases and controls over the first 6 weeks of life, after gestational adjustment. When combining with birth weight, the derived metabolic-based discriminative model observed some differences between those with and without BPD development, with c-statistics of 0.869 and 0.841 at day 7 and 28 of life on the test data. Conclusions: The metabolic panel we describe identified some metabolic differences in the blood associated with BPD pathogenesis. Further work is needed to determine whether these compounds could facilitate the monitoring and/or investigation of early-life metabolic status in the lung and other tissues for the prevention and management of BPD.
Collapse
Affiliation(s)
- Chengyin Ye
- Department of Health Management, School of Public Health, Hangzhou Normal University, Hangzhou 311100, China
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Jinghua Wu
- Department of Health Management, School of Public Health, Hangzhou Normal University, Hangzhou 311100, China
| | - Jonathan D. Reiss
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
- Stanford Metabolic Health Center, Stanford Children’s Hospital, Stanford, CA 94304, USA
| | - Tiffany J. Sinclair
- Department of Surgery, Division of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - David K. Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
- Stanford Metabolic Health Center, Stanford Children’s Hospital, Stanford, CA 94304, USA
| | - Gary M. Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | | | - Reese H. Clark
- Pediatrix-Obstetrix Center for Research, Education and Quality, Sunrise, FL 33323, USA
| | - Lawrence S. Prince
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Xuefeng Bruce Ling
- Department of Surgery, Division of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA
- Clinical and Translational Research Program, Betty Irene Moore Children’s Heart Center, Lucile Packard Children’s Hospital, Palo Alto, CA 94304, USA
- Correspondence: (X.B.L.); (K.G.S.); Tel.: +1-650-723-6439 (K.G.S.); Fax: +1-650-725-5577 (K.G.S.)
| | - Karl G. Sylvester
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
- Stanford Metabolic Health Center, Stanford Children’s Hospital, Stanford, CA 94304, USA
- Department of Surgery, Division of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA
- Correspondence: (X.B.L.); (K.G.S.); Tel.: +1-650-723-6439 (K.G.S.); Fax: +1-650-725-5577 (K.G.S.)
| |
Collapse
|
19
|
Abstract
CPT2 K79 acetylation caused by NAD+ exhaustion and Sirt3 dysfunction resulted in LCAC accumulation and platelet damage. Blocking acylcarnitine generation with AMPK or CPT1 inhibitors, Sirt3 agonists, and antioxidants retarded platelet storage lesion.
The short life span of platelets is a major challenge to platelet transfusion services because of the lack of effective intervention. Here, we found that the accumulation of long-chain acylcarnitines (LCACs) is responsible for mitochondrial damage and platelet storage lesion. Further studies showed that the blockade of fatty acid oxidation and the activation of AMP-activated protein kinase (AMPK)/acetyl-CoA carboxylase/carnitine palmitoyltransferase 1 (CPT1) pathways that promote fatty acid metabolism are important reasons for the accumulation of LCACs. The excessive accumulation of LCACs can cause mitochondrial damage and a short life span of stored platelets. The mechanism study elucidated that NAD+ exhaustion and the subsequent decrease in sirtuin 3 (Sirt3) activity caused an increase in the level of CPT2 K79 acetylation, which is the primary cause of the blockade of fatty acid oxidation and the accumulation of LCACs. Blocking LCAC generation with the inhibitors of AMPK or CPT1, the agonists of Sirt3, and antioxidants tremendously retarded platelet storage lesion in vitro and prolonged the survival of stored platelets in vivo posttransfusion with single or combined use. In summary, we discovered that CPT2 acetylation attenuates fatty acid oxidation and exacerbates platelet storage lesion and may serve as a new target for improving platelet storage quality.
Collapse
|
20
|
Liang L, Hu M, Chen Y, Liu L, Wu L, Hang C, Luo X, Xu X. Metabolomics of bronchoalveolar lavage in children with persistent wheezing. Respir Res 2022; 23:161. [PMID: 35718784 PMCID: PMC9208141 DOI: 10.1186/s12931-022-02087-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/11/2022] [Indexed: 11/28/2022] Open
Abstract
Background Recent studies have demonstrated the important role of metabolomics in the pathogenesis of asthma. However, the role of lung metabolomics in childhood persistent wheezing (PW) or wheezing recurrence remains poorly understood. Methods In this prospective observational study, we performed a liquid chromatography/mass spectrometry-based metabolomic survey on bronchoalveolar lavage samples collected from 30 children with PW and 30 age-matched infants (control group). A 2-year follow-up study on these PW children was conducted. Results Children with PW showed a distinct characterization of respiratory metabolome compared with control group. Children with PW had higher abundances of choline, oleamide, nepetalactam, butyrylcarnitine, l-palmitoylcarnitine, palmitoylethanolamide, and various phosphatidylcholines. The glycerophospholipid metabolism pathway was the most relevant pathway involving in PW pathophysiologic process. Additionally, different gender, prematurity, and systemic corticoids use demonstrated a greater impact in airway metabolite compositions. Furthermore, for PW children with recurrence during the follow-up period, children who were born prematurely had an increased abundance of butyrylcarnitine relative to those who were carried to term. Conclusions This study suggests that the alterations of lung metabolites could be associated with the development of wheezing, and this early alteration could also be correlated with wheezing recurrence later in life. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02087-6.
Collapse
Affiliation(s)
- Lingfang Liang
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Minfei Hu
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Yuanling Chen
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Lingke Liu
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Lei Wu
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Chengcheng Hang
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Xiaofei Luo
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Xuefeng Xu
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
21
|
Castañé H, Iftimie S, Baiges-Gaya G, Rodríguez-Tomàs E, Jiménez-Franco A, López-Azcona AF, Garrido P, Castro A, Camps J, Joven J. Machine learning and semi-targeted lipidomics identify distinct serum lipid signatures in hospitalized COVID-19-positive and COVID-19-negative patients. Metabolism 2022; 131:155197. [PMID: 35381232 PMCID: PMC8976580 DOI: 10.1016/j.metabol.2022.155197] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/11/2022] [Accepted: 03/28/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND Lipids are involved in the interaction between viral infection and the host metabolic and immunological responses. Several studies comparing the lipidome of COVID-19-positive hospitalized patients vs. healthy subjects have already been reported. It is largely unknown, however, whether these differences are specific to this disease. The present study compared the lipidomic signature of hospitalized COVID-19-positive patients with that of healthy subjects, as well as with COVID-19-negative patients hospitalized for other infectious/inflammatory diseases. METHODS We analyzed the lipidomic signature of 126 COVID-19-positive patients, 45 COVID-19-negative patients hospitalized with other infectious/inflammatory diseases and 50 healthy volunteers. A semi-targeted lipidomics analysis was performed using liquid chromatography coupled to mass spectrometry. Two-hundred and eighty-three lipid species were identified and quantified. Results were interpreted by machine learning tools. RESULTS We identified acylcarnitines, lysophosphatidylethanolamines, arachidonic acid and oxylipins as the most altered species in COVID-19-positive patients compared to healthy volunteers. However, we found similar alterations in COVID-19-negative patients who had other causes of inflammation. Conversely, lysophosphatidylcholine 22:6-sn2, phosphatidylcholine 36:1 and secondary bile acids were the parameters that had the greatest capacity to discriminate between COVID-19-positive and COVID-19-negative patients. CONCLUSION This study shows that COVID-19 infection shares many lipid alterations with other infectious/inflammatory diseases, and which differentiate them from the healthy population. The most notable alterations were observed in oxylipins, while alterations in bile acids and glycerophospholipis best distinguished between COVID-19-positive and COVID-19-negative patients. Our results highlight the value of integrating lipidomics with machine learning algorithms to explore the pathophysiology of COVID-19 and, consequently, improve clinical decision making.
Collapse
Affiliation(s)
- Helena Castañé
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Simona Iftimie
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Gerard Baiges-Gaya
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Elisabet Rodríguez-Tomàs
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Andrea Jiménez-Franco
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Ana Felisa López-Azcona
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Pedro Garrido
- Intensive Care Unit, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Antoni Castro
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain.
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
22
|
Hussain H, Vutipongsatorn K, Jiménez B, Antcliffe DB. Patient Stratification in Sepsis: Using Metabolomics to Detect Clinical Phenotypes, Sub-Phenotypes and Therapeutic Response. Metabolites 2022; 12:metabo12050376. [PMID: 35629881 PMCID: PMC9145582 DOI: 10.3390/metabo12050376] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/01/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
Infections are common and need minimal treatment; however, occasionally, due to inappropriate immune response, they can develop into a life-threatening condition known as sepsis. Sepsis is a global concern with high morbidity and mortality. There has been little advancement in the treatment of sepsis, outside of antibiotics and supportive measures. Some of the difficulty in identifying novel therapies is the heterogeneity of the condition. Metabolic phenotyping has great potential for gaining understanding of this heterogeneity and how the metabolic fingerprints of patients with sepsis differ based on survival, organ dysfunction, disease severity, type of infection, treatment or causative organism. Moreover, metabolomics offers potential for patient stratification as metabolic profiles obtained from analytical platforms can reflect human individuality and phenotypic variation. This article reviews the most relevant metabolomic studies in sepsis and aims to provide an overview of the metabolic derangements in sepsis and how metabolic phenotyping has been used to identify sub-groups of patients with this condition. Finally, we consider the new avenues that metabolomics could open, exploring novel phenotypes and untangling the heterogeneity of sepsis, by looking at advances made in the field with other -omics technologies.
Collapse
Affiliation(s)
- Humma Hussain
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK; (H.H.); (K.V.)
| | - Kritchai Vutipongsatorn
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK; (H.H.); (K.V.)
| | - Beatriz Jiménez
- Section of Bioanalytical Chemistry, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK;
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - David B. Antcliffe
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK; (H.H.); (K.V.)
- Correspondence:
| |
Collapse
|
23
|
Valdés A, Moreno LO, Rello SR, Orduña A, Bernardo D, Cifuentes A. Metabolomics study of COVID-19 patients in four different clinical stages. Sci Rep 2022; 12:1650. [PMID: 35102215 PMCID: PMC8803913 DOI: 10.1038/s41598-022-05667-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 12/13/2021] [Indexed: 12/22/2022] Open
Abstract
SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is the coronavirus strain causing the respiratory pandemic COVID-19 (coronavirus disease 2019). To understand the pathobiology of SARS-CoV-2 in humans it is necessary to unravel the metabolic changes that are produced in the individuals once the infection has taken place. The goal of this work is to provide new information about the altered biomolecule profile and with that the altered biological pathways of patients in different clinical situations due to SARS-CoV-2 infection. This is done via metabolomics using HPLC-QTOF-MS analysis of plasma samples at COVID-diagnose from a total of 145 adult patients, divided into different clinical stages based on their subsequent clinical outcome (25 negative controls (non-COVID); 28 positive patients with asymptomatic disease not requiring hospitalization; 27 positive patients with mild disease defined by a total time in hospital lower than 10 days; 36 positive patients with severe disease defined by a total time in hospital over 20 days and/or admission at the ICU; and 29 positive patients with fatal outcome or deceased). Moreover, follow up samples between 2 and 3 months after hospital discharge were also obtained from the hospitalized patients with mild prognosis. The final goal of this work is to provide biomarkers that can help to better understand how the COVID-19 illness evolves and to predict how a patient could progress based on the metabolites profile of plasma obtained at an early stage of the infection. In the present work, several metabolites were found as potential biomarkers to distinguish between the end-stage and the early-stage (or non-COVID) disease groups. These metabolites are mainly involved in the metabolism of carnitines, ketone bodies, fatty acids, lysophosphatidylcholines/phosphatidylcholines, tryptophan, bile acids and purines, but also omeprazole. In addition, the levels of several of these metabolites decreased to "normal" values at hospital discharge, suggesting some of them as early prognosis biomarkers in COVID-19 at diagnose.
Collapse
Affiliation(s)
- Alberto Valdés
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolás Cabrera 9, 28049, Madrid, Spain
| | - Lorena Ortega Moreno
- Dpt. Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Silvia Rojo Rello
- Servicio de Microbiología, Hospital Clínico Universitario de Valladolid, 47004, Valladolid, Spain
| | - Antonio Orduña
- Servicio de Microbiología, Hospital Clínico Universitario de Valladolid, 47004, Valladolid, Spain
- Departamento de Microbiología, Universidad de Valladolid, Valladolid, Spain
| | - David Bernardo
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Alejandro Cifuentes
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolás Cabrera 9, 28049, Madrid, Spain.
| |
Collapse
|
24
|
Thomas JM, Sudhadevi T, Basa P, Ha AW, Natarajan V, Harijith A. The Role of Sphingolipid Signaling in Oxidative Lung Injury and Pathogenesis of Bronchopulmonary Dysplasia. Int J Mol Sci 2022; 23:ijms23031254. [PMID: 35163176 PMCID: PMC8835774 DOI: 10.3390/ijms23031254] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Premature infants are born with developing lungs burdened by surfactant deficiency and a dearth of antioxidant defense systems. Survival rate of such infants has significantly improved due to advances in care involving mechanical ventilation and oxygen supplementation. However, a significant subset of such survivors develops the chronic lung disease, Bronchopulmonary dysplasia (BPD), characterized by enlarged, simplified alveoli and deformed airways. Among a host of factors contributing to the pathogenesis is oxidative damage induced by exposure of the developing lungs to hyperoxia. Recent data indicate that hyperoxia induces aberrant sphingolipid signaling, leading to mitochondrial dysfunction and abnormal reactive oxygen species (ROS) formation (ROS). The role of sphingolipids such as ceramides and sphingosine 1-phosphate (S1P), in the development of BPD emerged in the last decade. Both ceramide and S1P are elevated in tracheal aspirates of premature infants of <32 weeks gestational age developing BPD. This was faithfully reflected in the murine models of hyperoxia and BPD, where there is an increased expression of sphingolipid metabolites both in lung tissue and bronchoalveolar lavage. Treatment of neonatal pups with a sphingosine kinase1 specific inhibitor, PF543, resulted in protection against BPD as neonates, accompanied by improved lung function and reduced airway remodeling as adults. This was accompanied by reduced mitochondrial ROS formation. S1P receptor1 induced by hyperoxia also aggravates BPD, revealing another potential druggable target in this pathway for BPD. In this review we aim to provide a detailed description on the role played by sphingolipid signaling in hyperoxia induced lung injury and BPD.
Collapse
Affiliation(s)
- Jaya M. Thomas
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Tara Sudhadevi
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Prathima Basa
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Alison W. Ha
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Viswanathan Natarajan
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Anantha Harijith
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
- Correspondence: ; Tel.: +1-(216)-286-7038
| |
Collapse
|
25
|
van Liempd S, Cabrera D, Pilzner C, Kollmus H, Schughart K, Falcón-Pérez JM. Impaired beta-oxidation increases vulnerability to influenza A infection. J Biol Chem 2021; 297:101298. [PMID: 34637789 PMCID: PMC8564733 DOI: 10.1016/j.jbc.2021.101298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/20/2022] Open
Abstract
Influenza A virus (IAV) infection casts a significant burden on society. It has particularly high morbidity and mortality rates in patients suffering from metabolic disorders. The aim of this study was to relate metabolic changes with IAV susceptibility using well-characterized inbred mouse models. We compared the highly susceptible DBA/2J (D2) mouse strain for which IAV infection is lethal with the C57BL/6J (B6) strain, which exhibits a moderate course of disease and survives IAV infection. Previous studies showed that D2 has higher insulin and glucose levels and is predisposed to develop diet-induced type 2 diabetes. Using high-resolution liquid chromatography–coupled MS, the plasma metabolomes of individual animals were repeatedly measured up to 30 days postinfection. The biggest metabolic difference between these strains in healthy and infected states was in the levels of malonylcarnitine, which was consistently increased 5-fold in D2. Other interstrain and intrastrain differences in healthy and infected animals were observed for acylcarnitines, glucose, branched-chain amino acids, and oxidized fatty acids. By mapping metabolic changes to canonical pathways, we found that mitochondrial beta-oxidation is likely disturbed in D2 animals. In noninfected D2 mice, this leads to increased glycerolipid production and reduced acylcarnitine production, whereas in infected D2 animals, peroxisomal beta-oxidation becomes strongly increased. From these studies, we conclude that metabolic changes caused by a distortion of mitochondrial and peroxisomal metabolism might impact the innate immune response in D2, leading to high viral titers and mortality.
Collapse
Affiliation(s)
| | - Diana Cabrera
- Metabolomics Platform CIC bioGUNE-BRTA, Derio, Spain
| | - Carolin Pilzner
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Heike Kollmus
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany; University of Veterinary Medicine Hannover, Hannover, Germany; Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Juan M Falcón-Pérez
- Metabolomics Platform CIC bioGUNE-BRTA, Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
26
|
Mussap M, Fanos V. Could metabolomics drive the fate of COVID-19 pandemic? A narrative review on lights and shadows. Clin Chem Lab Med 2021; 59:1891-1905. [PMID: 34332518 DOI: 10.1515/cclm-2021-0414] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Human Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) infection activates a complex interaction host/virus, leading to the reprogramming of the host metabolism aimed at the energy supply for viral replication. Alterations of the host metabolic homeostasis strongly influence the immune response to SARS-CoV-2, forming the basis of a wide range of outcomes, from the asymptomatic infection to the onset of COVID-19 and up to life-threatening acute respiratory distress syndrome, vascular dysfunction, multiple organ failure, and death. Deciphering the molecular mechanisms associated with the individual susceptibility to SARS-CoV-2 infection calls for a system biology approach; this strategy can address multiple goals, including which patients will respond effectively to the therapeutic treatment. The power of metabolomics lies in the ability to recognize endogenous and exogenous metabolites within a biological sample, measuring their concentration, and identifying perturbations of biochemical pathways associated with qualitative and quantitative metabolic changes. Over the last year, a limited number of metabolomics- and lipidomics-based clinical studies in COVID-19 patients have been published and are discussed in this review. Remarkable alterations in the lipid and amino acid metabolism depict the molecular phenotype of subjects infected by SARS-CoV-2; notably, structural and functional data on the lipids-virus interaction may open new perspectives on targeted therapeutic interventions. Several limitations affect most metabolomics-based studies, slowing the routine application of metabolomics. However, moving metabolomics from bench to bedside cannot imply the mere determination of a given metabolite panel; rather, slotting metabolomics into clinical practice requires the conversion of metabolic patient-specific data into actionable clinical applications.
Collapse
Affiliation(s)
- Michele Mussap
- Laboratory Medicine, Department of Surgical Sciences, School of Medicine, University of Cagliari, Monserrato, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, School of Medicine, University of Cagliari, Monserrato, Italy
| |
Collapse
|
27
|
Hepokoski M, Wang J, Li K, Li Y, Gupta P, Mai T, Moshensky A, Alotaibi M, Crotty Alexander LE, Malhotra A, Singh P. Altered lung metabolism and mitochondrial DAMPs in lung injury due to acute kidney injury. Am J Physiol Lung Cell Mol Physiol 2021; 320:L821-L831. [PMID: 33565357 PMCID: PMC8174821 DOI: 10.1152/ajplung.00578.2020] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common cause of mortality in patients with acute kidney injury (AKI). Inflammatory crosstalk from the kidney to the lung has been shown to contribute to lung injury after AKI, but anti-inflammatory therapies have not been proven beneficial in human studies. Recently, AKI was shown to alter mitochondria and related metabolic pathways in the heart, but the impact of AKI on lung metabolism has not been investigated to our knowledge. In this study, we evaluated the metabolomic profile of the lung following renal ischemia and reperfusion to identify novel pathways that may be modifiable. We randomized C57BL/6 mice to 20 minutes of bilateral renal arterial clamping or sham operation under ketamine/xylazine anesthesia. At 4 hours after reperfusion, we found a significant increase in markers of lung injury, as well as significant metabolomic changes across lung, kidney, plasma and bronchoalveolar lavage fluid (BALF) compared to shams. Comparative analyses revealed that the fatty acid oxidation pathway was the most significantly altered metabolic pathway, a finding which is consistent with mitochondrial dysfunction systemically and in the lung. These metabolomic changes correlated with the extracellular accumulation of the mitochondrial damage associated molecular patterns (mtDAMPs), mitochondrial DNA (mtDNA) and transcription factor A, mitochondria (TFAM). Finally, we found that intraperitoneal injection of renal mtDAMPs caused metabolomic changes consistent with mitochondrial dysfunction in the lung in vivo. Mitochondrial function and mtDAMPs warrant further investigation as potential therapeutic targets in preventing lung injury because of AKI.
Collapse
Affiliation(s)
- Mark Hepokoski
- 1VA San Diego Healthcare System, San Diego, California,2Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, California,4Department of Medicine, School of Medicine, University of California, San Diego, California
| | - Jing Wang
- 2Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, California,4Department of Medicine, School of Medicine, University of California, San Diego, California,5Department of Critical Care Medicine, Yantai Yuhuangding Hospital, Affiliated with Medical College of Qingdao University, Yantai, Shangdong, China
| | - Kefeng Li
- 4Department of Medicine, School of Medicine, University of California, San Diego, California
| | - Ying Li
- 1VA San Diego Healthcare System, San Diego, California,3Division of Nephrology and Hypertension, University of California San Diego, California,4Department of Medicine, School of Medicine, University of California, San Diego, California
| | - Purva Gupta
- 1VA San Diego Healthcare System, San Diego, California,2Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, California,4Department of Medicine, School of Medicine, University of California, San Diego, California
| | - Tina Mai
- 1VA San Diego Healthcare System, San Diego, California
| | - Alex Moshensky
- 1VA San Diego Healthcare System, San Diego, California,2Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, California,4Department of Medicine, School of Medicine, University of California, San Diego, California
| | - Mona Alotaibi
- 1VA San Diego Healthcare System, San Diego, California,2Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, California,4Department of Medicine, School of Medicine, University of California, San Diego, California
| | - Laura E. Crotty Alexander
- 1VA San Diego Healthcare System, San Diego, California,2Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, California,4Department of Medicine, School of Medicine, University of California, San Diego, California
| | - Atul Malhotra
- 2Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, California,4Department of Medicine, School of Medicine, University of California, San Diego, California
| | - Prabhleen Singh
- 1VA San Diego Healthcare System, San Diego, California,3Division of Nephrology and Hypertension, University of California San Diego, California,4Department of Medicine, School of Medicine, University of California, San Diego, California
| |
Collapse
|
28
|
Xuefei Y, Xinyi Z, Qing C, Dan Z, Ziyun L, Hejuan Z, Xindong X, Jianhua F. Effects of Hyperoxia on Mitochondrial Homeostasis: Are Mitochondria the Hub for Bronchopulmonary Dysplasia? Front Cell Dev Biol 2021; 9:642717. [PMID: 33996802 PMCID: PMC8120003 DOI: 10.3389/fcell.2021.642717] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/12/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondria are involved in energy metabolism and redox reactions in the cell. Emerging data indicate that mitochondria play an essential role in physiological and pathological processes of neonatal lung development. Mitochondrial damage due to exposure to high concentrations of oxygen is an indeed important factor for simplification of lung structure and development of bronchopulmonary dysplasia (BPD), as reported in humans and rodent models. Here, we comprehensively review research that have determined the effects of oxygen environment on alveolar development and morphology, summarize changes in mitochondria under high oxygen concentrations, and discuss several mitochondrial mechanisms that may affect cell plasticity and their effects on BPD. Thus, the pathophysiological effects of mitochondria may provide insights into targeted mitochondrial and BPD therapy.
Collapse
Affiliation(s)
- Yu Xuefei
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Zhao Xinyi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Cai Qing
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Zhang Dan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Liu Ziyun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Zheng Hejuan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Xue Xindong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Fu Jianhua
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| |
Collapse
|
29
|
OUYANG Y, CHI L, XU C, ZHAO X, CUI Z. [Liquid chromatography-mass spectrometry-based metabolomics study of the efficacy of Chinese medicine asthma-relieving decoction on respiratory syncytial virus infection]. Se Pu 2021; 39:281-290. [PMID: 34227309 PMCID: PMC9403810 DOI: 10.3724/sp.j.1123.2020.06013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Indexed: 11/25/2022] Open
Abstract
Respiratory syncytial virus (RSV) can cause lower respiratory tract infections, such as bronchiolitis in infants. In China, traditional asthma-relieving medicine has numerous clinical applications in the treatment of RSV infections. However, due to the complexity of the traditional Chinese medicine system, its therapeutic mechanism and main pharmacological components remain unclear. Metabolomics can be used to analyze the efficacy of traditional Chinese medicine to provide modern scientific evidence for such treatments. In this study, an animal model experiment was performed with seven groups of three-week-old rats. The model group and five intervention groups were inoculated nasally with RSV for three consecutive days, and the normal group was treated with the same amount of saline for three consecutive days under the same conditions. In parallel, the five intervention groups were treated separately with the following via intragastric administration for seven consecutive days: asthma-relieving traditional Chinese medicine decoction, its three constituent agents (ascending (xuan) therapy, descending (jiang) therapy, pyretic clearing (qing) therapy), and ribavirin. Both normal group and RSV model group were administered with normal saline via intragastric administration as controls for seven consecutive days. The fundus plasma of rats in each group was collected on day 0, day 3, and day 7. Liquid chromatography-mass spectrometry-based untargeted metabolomics analysis was performed to investigate the changes in the metabolome after RSV infection, the effects of the asthma-relieving decoction on the regulation of metabolites related to RSV infection, and the primary source of efficacy. The detected metabolite ions were corrected using internal standards. Multivariate analysis of ions with an RSD value of less than 30% in quality control (QC) samples was used to construct principal component analysis models to monitor the overall metabolic changes of each group. The results showed that, during RSV infection and treatment, the asthma-relieving decoction and the positive control ribavirin had similar effects on the overall metabolic regulation of RSV-infected rats. Among the three asthma-relieving decoction constituent agents, the ascending (xuan) therapy agents which was composed of ephedra and ginkgo had a closer metabolic regulation effect with asthma-relieving decoction, and might be the main source of pharmacological efficacy. Based on the retention time, m/z value and tandem mass spectra in the database established by our laboratory, a total of 150 metabolites were identified. Paired t-tests were performed using data of the identified metabolites before and after RSV infection in each group, and it was found that 83 metabolite levels significantly changed after RSV infection, indicating that RSV infection could lead to disorders of multiple metabolic pathways in rats. The altered pathways included aminoacyl-tRNA biosynthesis, phenylalanine, tyrosine, and tryptophan biosynthesis, primary bile acid biosynthesis, phenylalanine metabolism and sphingomyelin metabolism. On the third day, the asthma-relieving decoction had regulatory effects on several metabolites such as bile acids, amino acids, organic acids, lipids, etc. Among the three asthma-relieving decoction constituent agents, the ascending (xuan) therapy agents had more similar effects on the regulation of metabolites with the asthma-relieving decoction. On the other hand, the descending (jiang) therapy agents and pyretic clearing (qing) therapy agents down-regulated the abnormal increase in acylcarnitine caused by the RSV infection. Additionally, both asthma-relieving decoction and its constituent agents could maintain the stability of the immune system and metabolism of the intestinal flora in rats. This study used metabolomics to evaluate the efficacy of an asthma-relieving decoction and demonstrate the metabolites and the corresponding changes after asthma-relieving decoction-based treatment. It provides theoretical support for research on the therapeutic mechanism and active ingredients of asthma-relieving decoction.
Collapse
|
30
|
Lee DD, Park SJ, Zborek KL, Schwarz MA. A shift from glycolytic and fatty acid derivatives toward one-carbon metabolites in the developing lung during transitions of the early postnatal period. Am J Physiol Lung Cell Mol Physiol 2021; 320:L640-L659. [PMID: 33502935 DOI: 10.1152/ajplung.00417.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
During postnatal lung development, metabolic changes that coincide with stages of alveolar formation are poorly understood. Responding to developmental and environmental factors, metabolic changes can be rapidly and adaptively altered. The objective of the present study was to determine biological and technical determinants of metabolic changes during postnatal lung development. Over 118 metabolic features were identified by liquid chromatography with tandem mass spectrometry (LC-MS/MS, Sciex QTRAP 5500 Triple Quadrupole). Biological determinants of metabolic changes were the transition from the postnatal saccular to alveolar stages and exposure to 85% hyperoxia, an environmental insult. Technical determinants of metabolic identification were brevity and temperature of harvesting, both of which improved metabolic preservation within samples. Multivariate statistical analyses revealed the transition between stages of lung development as the period of major metabolic alteration. Of three distinctive groups that clustered by age, the saccular stage was identified by its enrichment of both glycolytic and fatty acid derivatives. The critical transition between stages of development were denoted by changes in amino acid derivatives. Of the amino acid derivatives that significantly changed, a majority were linked to metabolites of the one-carbon metabolic pathway. The enrichment of one-carbon metabolites was independent of age and environmental insult. Temperature was also found to significantly influence the metabolic levels within the postmortem sampled lung, which underscored the importance of methodology. Collectively, these data support not only distinctive stages of metabolic change but also highlight amino acid metabolism, in particular one-carbon metabolites as metabolic signatures of the early postnatal lung.
Collapse
Affiliation(s)
- Daniel D Lee
- Department of Pediatrics, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana.,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | - Sang Jun Park
- Department of Preprofessional Studies, University of Notre Dame, South Bend, Indiana
| | - Kirsten L Zborek
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | - Margaret A Schwarz
- Department of Pediatrics, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana.,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| |
Collapse
|
31
|
Bei Y, Tia B, Li Y, Guo Y, Deng S, Huang R, Zeng H, Li R, Wang GF, Dai J. Anti-influenza A Virus Effects and Mechanisms of Emodin and Its Analogs via Regulating PPAR α/ γ-AMPK-SIRT1 Pathway and Fatty Acid Metabolism. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9066938. [PMID: 34540999 PMCID: PMC8445710 DOI: 10.1155/2021/9066938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/14/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023]
Abstract
The peroxisome proliferator-activated receptor (PPAR) α/γ-adenosine 5'-monophosphate- (AMP-) activated protein kinase- (AMPK-) sirtuin-1 (SIRT1) pathway and fatty acid metabolism are reported to be involved in influenza A virus (IAV) replication and IAV-pneumonia. Through a cell-based peroxisome proliferator responsive element- (PPRE-) driven luciferase bioassay, we have investigated 145 examples of traditional Chinese medicines (TCMs). Several TCMs, such as Polygonum cuspidatum, Rheum officinale Baillon, and Aloe vera var. Chinensis (Haw.) Berg., were found to possess high activity. We have further detected the anti-IAV activities of emodin (EMO) and its analogs, a group of common important compounds of these TCMs. The results showed that emodin and its several analogs possess excellent anti-IAV activities. The pharmacological tests showed that emodin significantly activated PPARα/γ and AMPK, decreased fatty acid biosynthesis, and increased intracellular ATP levels. Pharmaceutical inhibitors, siRNAs for PPARα/γ and AMPKα1, and exogenous palmitate impaired the inhibition of emodin. The in vivo test also showed that emodin significantly protected mice from IAV infection and pneumonia. Pharmacological inhibitors for PPARα/γ and AMPK signal and exogenous palmitate could partially counteract the effects of emodin in vivo. In conclusion, emodin and its analogs are a group of promising anti-IAV drug precursors, and the pharmacological mechanism of emodin is linked to its ability to regulate the PPARα/γ-AMPK pathway and fatty acid metabolism.
Collapse
Affiliation(s)
- Yufei Bei
- Department of Pharmacy, Affiliated Hospital of Nantong University, 20th Xisi Road, 226 001 Nantong, China
- Department of Microbiology and Immunology, Shantou University Medical College, Xinling Road, 22, Shantou, Guangdong 515 041, China
| | - Boyu Tia
- Department of Microbiology and Immunology, Shantou University Medical College, Xinling Road, 22, Shantou, Guangdong 515 041, China
| | - Yuze Li
- Department of Microbiology and Immunology, Shantou University Medical College, Xinling Road, 22, Shantou, Guangdong 515 041, China
| | - Yingzhu Guo
- Department of Microbiology and Immunology, Shantou University Medical College, Xinling Road, 22, Shantou, Guangdong 515 041, China
| | - Shufei Deng
- Department of Microbiology and Immunology, Shantou University Medical College, Xinling Road, 22, Shantou, Guangdong 515 041, China
| | - Rouyu Huang
- Department of Microbiology and Immunology, Shantou University Medical College, Xinling Road, 22, Shantou, Guangdong 515 041, China
| | - Huiling Zeng
- Department of Microbiology and Immunology, Shantou University Medical College, Xinling Road, 22, Shantou, Guangdong 515 041, China
| | - Rui Li
- Department of Microbiology and Immunology, Shantou University Medical College, Xinling Road, 22, Shantou, Guangdong 515 041, China
| | - Ge-Fei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Xinling Road, 22, Shantou, Guangdong 515 041, China
| | - Jianping Dai
- Department of Microbiology and Immunology, Shantou University Medical College, Xinling Road, 22, Shantou, Guangdong 515 041, China
| |
Collapse
|
32
|
Organic Cation Transporters in the Lung-Current and Emerging (Patho)Physiological and Pharmacological Concepts. Int J Mol Sci 2020; 21:ijms21239168. [PMID: 33271927 PMCID: PMC7730617 DOI: 10.3390/ijms21239168] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Organic cation transporters (OCT) 1, 2 and 3 and novel organic cation transporters (OCTN) 1 and 2 of the solute carrier 22 (SLC22) family are involved in the cellular transport of endogenous compounds such as neurotransmitters, l-carnitine and ergothioneine. OCT/Ns have also been implicated in the transport of xenobiotics across various biological barriers, for example biguanides and histamine receptor antagonists. In addition, several drugs used in the treatment of respiratory disorders are cations at physiological pH and potential substrates of OCT/Ns. OCT/Ns may also be associated with the development of chronic lung diseases such as allergic asthma and chronic obstructive pulmonary disease (COPD) and, thus, are possible new drug targets. As part of the Special Issue "Physiology, Biochemistry and Pharmacology of Transporters for Organic Cations", this review provides an overview of recent findings on the (patho)physiological and pharmacological functions of organic cation transporters in the lung.
Collapse
|
33
|
Barberis E, Timo S, Amede E, Vanella VV, Puricelli C, Cappellano G, Raineri D, Cittone MG, Rizzi E, Pedrinelli AR, Vassia V, Casciaro FG, Priora S, Nerici I, Galbiati A, Hayden E, Falasca M, Vaschetto R, Sainaghi PP, Dianzani U, Rolla R, Chiocchetti A, Baldanzi G, Marengo E, Manfredi M. Large-Scale Plasma Analysis Revealed New Mechanisms and Molecules Associated with the Host Response to SARS-CoV-2. Int J Mol Sci 2020; 21:E8623. [PMID: 33207699 PMCID: PMC7696386 DOI: 10.3390/ijms21228623] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 01/08/2023] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread to nearly every continent, registering over 1,250,000 deaths worldwide. The effects of SARS-CoV-2 on host targets remains largely limited, hampering our understanding of Coronavirus Disease 2019 (COVID-19) pathogenesis and the development of therapeutic strategies. The present study used a comprehensive untargeted metabolomic and lipidomic approach to capture the host response to SARS-CoV-2 infection. We found that several circulating lipids acted as potential biomarkers, such as phosphatidylcholine 14:0_22:6 (area under the curve (AUC) = 0.96), phosphatidylcholine 16:1_22:6 (AUC = 0.97), and phosphatidylethanolamine 18:1_20:4 (AUC = 0.94). Furthermore, triglycerides and free fatty acids, especially arachidonic acid (AUC = 0.99) and oleic acid (AUC = 0.98), were well correlated to the severity of the disease. An untargeted analysis of non-critical COVID-19 patients identified a strong alteration of lipids and a perturbation of phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine metabolism, aminoacyl-tRNA degradation, arachidonic acid metabolism, and the tricarboxylic acid (TCA) cycle. The severity of the disease was characterized by the activation of gluconeogenesis and the metabolism of porphyrins, which play a crucial role in the progress of the infection. In addition, our study provided further evidence for considering phospholipase A2 (PLA2) activity as a potential key factor in the pathogenesis of COVID-19 and a possible therapeutic target. To date, the present study provides the largest untargeted metabolomics and lipidomics analysis of plasma from COVID-19 patients and control groups, identifying new mechanisms associated with the host response to COVID-19, potential plasma biomarkers, and therapeutic targets.
Collapse
Affiliation(s)
- Elettra Barberis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
| | - Sara Timo
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Alessandria, Italy
| | - Elia Amede
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
| | - Virginia V. Vanella
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
| | - Chiara Puricelli
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Giuseppe Cappellano
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Davide Raineri
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Micol G. Cittone
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Eleonora Rizzi
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Anita R. Pedrinelli
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Veronica Vassia
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Francesco G. Casciaro
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Simona Priora
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Ilaria Nerici
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Alessandra Galbiati
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Eyal Hayden
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin University, Perth 6102, Australia;
| | - Rosanna Vaschetto
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
| | - Pier Paolo Sainaghi
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Roberta Rolla
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Annalisa Chiocchetti
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Gianluca Baldanzi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
| | - Emilio Marengo
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Alessandria, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
| |
Collapse
|
34
|
Beck ME, Zhang Y, Bharathi SS, Kosmider B, Bahmed K, Dahmer MK, Nogee LM, Goetzman ES. The common K333Q polymorphism in long-chain acyl-CoA dehydrogenase (LCAD) reduces enzyme stability and function. Mol Genet Metab 2020; 131:83-89. [PMID: 32389575 PMCID: PMC7606262 DOI: 10.1016/j.ymgme.2020.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 01/19/2023]
Abstract
The fatty acid oxidation enzyme long-chain acyl-CoA dehydrogenase (LCAD) is expressed at high levels in human alveolar type II (ATII) cells in the lung. A common polymorphism causing an amino acid substitution (K333Q) was previously linked to a loss of LCAD antigen in the lung tissue in sudden infant death syndrome. However, the effects of the polymorphism on LCAD function has not been tested. The present work evaluated recombinant LCAD K333Q. Compared to wild-type LCAD protein, LCAD K333Q exhibited significantly reduced enzymatic activity. Molecular modeling suggested that K333 is within interacting distance of the essential FAD cofactor, and the K333Q protein showed a propensity to lose FAD. Exogenous FAD only partially rescued the activity of LCAD K333Q. LCAD K333Q protein was less stable than wild-type when incubated at physiological temperatures, likely explaining the observation of dramatically reduced LCAD antigen in primary ATII cells isolated from individuals homozygous for K333Q. Despite the effect of K333Q on activity, stability, and antigen levels, the frequency of the polymorphism was not increased among infants and children with lung disease.
Collapse
Affiliation(s)
- Megan E Beck
- Department of Pediatrics, Division of Medical Genetics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, United States of America
| | - Yuxun Zhang
- Department of Pediatrics, Division of Medical Genetics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, United States of America
| | - Sivakama S Bharathi
- Department of Pediatrics, Division of Medical Genetics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, United States of America
| | - Beata Kosmider
- Department of Physiology, Temple University, Philadelphia, PA 19140, United States of America; Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, United States of America; Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, PA 19140, United States of America; Department of Medicine, National Jewish Health, Denver, CO 80206, United States of America
| | - Karim Bahmed
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, United States of America; Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, PA 19140, United States of America
| | - Mary K Dahmer
- Department of Pediatrics, Division of Critical Care, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Lawrence M Nogee
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States of America
| | - Eric S Goetzman
- Department of Pediatrics, Division of Medical Genetics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, United States of America.
| |
Collapse
|
35
|
Bannuscher A, Hellack B, Bahl A, Laloy J, Herman H, Stan MS, Dinischiotu A, Giusti A, Krause BC, Tentschert J, Roșu M, Balta C, Hermenean A, Wiemann M, Luch A, Haase A. Metabolomics profiling to investigate nanomaterial toxicity in vitro and in vivo. Nanotoxicology 2020; 14:807-826. [DOI: 10.1080/17435390.2020.1764123] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Anne Bannuscher
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
- Adolphe Merkle Institute (AMI), University of Fribourg, Fribourg, Switzerland
| | - Bryan Hellack
- Institute of Energy and Environmental Technology (IUTA) e.V, Duisburg, Germany
- German Environment Agency (UBA), Dessau, Germany
| | - Aileen Bahl
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Julie Laloy
- Department of Pharmacy, Namur Nanosafety Centre, NARILIS, University of Namur, Namur, Belgium
| | - Hildegard Herman
- Aurel Ardelean” Institute of Life Sciences, “Vasile Goldis” Western University of Arad, Arad, Romania
| | - Miruna S. Stan
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Anna Giusti
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Benjamin-Christoph Krause
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Jutta Tentschert
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Marcel Roșu
- Aurel Ardelean” Institute of Life Sciences, “Vasile Goldis” Western University of Arad, Arad, Romania
| | - Cornel Balta
- Aurel Ardelean” Institute of Life Sciences, “Vasile Goldis” Western University of Arad, Arad, Romania
| | - Anca Hermenean
- Aurel Ardelean” Institute of Life Sciences, “Vasile Goldis” Western University of Arad, Arad, Romania
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Martin Wiemann
- IBE R&D Institute for Lung Health gGmbH, Münster, Germany
| | - Andreas Luch
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Andrea Haase
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| |
Collapse
|
36
|
Jargin SV. Scientific Papers and Patents on Substances with Unproven Effects. ACTA ACUST UNITED AC 2020; 13:37-45. [PMID: 30848224 DOI: 10.2174/1872211313666190307162041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/26/2019] [Accepted: 02/26/2019] [Indexed: 01/22/2023]
Abstract
It is evident from reviewing scientific literature that the quality of argumentation in some areas of medical research has deteriorated during the last decades. Publication of a series of questionable reliability has continued without making references to the published criticism; examples are discussed in this review. Another tendency is that drugs without proven efficiency are advertised, corresponding products patented and marketed as evidence-based medications. Professional publications are required to register drugs and dietary supplements to obtain permissions for the practical use; and such papers appeared, sometimes being of questionable reliability. Several examples are discussed in this review when substances without proven effects were patented and introduced into practice being supported by publications of questionable reliability. Some of the topics are not entirely clear; and the arguments provided here can induce a constructive discussion.
Collapse
Affiliation(s)
- Sergei V Jargin
- Peoples' Friendship University of Russia, Clementovski per 6-82, 115184 Moscow, Russian Federation
| |
Collapse
|
37
|
Anwer W, Ratto Velasquez A, Tsoukanova V. Acylcarnitines at the Membrane Surface: Insertion Parameters for a Mitochondrial Leaflet Model. Biophys J 2020; 118:1032-1043. [PMID: 32027823 DOI: 10.1016/j.bpj.2020.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/11/2019] [Accepted: 01/14/2020] [Indexed: 12/28/2022] Open
Abstract
Excessive accumulation of acylcarnitines (ACs), often caused by metabolic disorders, has been associated with obesity, arrhythmias, cardiac ischemia, insulin resistance, etc. Mechanisms whereby elevated ACs might contribute to pathophysiological effects remain largely unexplored. We have aimed to gain insight into AC interactions with the mitochondrial inner membrane. To model its outer leaflet, Langmuir monolayers and cushioned supported bilayers were employed. Their interactions with ACs were monitored with epifluorescence microscopy, which revealed a local leaflet expansion upon exposure to elevated concentrations of a long-chain AC, plausibly caused by its insertion. To assess the AC insertion parameters, constant-pressure insertion assays were performed. A value of 21 ± 3 Å2 was obtained for the AC insertion area, which is roughly the same as the cross-sectional area of an acyl chain. By contrast, the carnitine moiety was found to require an area of 37 ± 3 Å2. The AC insertion has thus been concluded to involve solely the AC acyl chain. This mode of insertion implies that the carnitine moiety, with its nontitratable positive charge, is left dangling at the membrane surface, which is likely to alter the surface electrostatics of the outer leaflet. The extrapolation of these findings has enabled us to hypothesize that, by altering the morphology and surface electrostatics of the outer leaflet, the insertion of ACs, in particular their long-chain counterparts, may trigger a nonspecific activation of signaling pathways in the inner mitochondrial membrane, thereby modulating its function and potentially leading to pathophysiological responses.
Collapse
Affiliation(s)
- Wajih Anwer
- Department of Chemistry, York University, Toronto, Ontario, Canada
| | | | | |
Collapse
|
38
|
Yang H, Zhao C, Tang MC, Wang Y, Wang SP, Allard P, Furtos A, Mitchell GA. Inborn errors of mitochondrial acyl-coenzyme a metabolism: acyl-CoA biology meets the clinic. Mol Genet Metab 2019; 128:30-44. [PMID: 31186158 DOI: 10.1016/j.ymgme.2019.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/30/2019] [Accepted: 05/05/2019] [Indexed: 12/18/2022]
Abstract
The last decade saw major advances in understanding the metabolism of Coenzyme A (CoA) thioesters (acyl-CoAs) and related inborn errors (CoA metabolic diseases, CAMDs). For diagnosis, acylcarnitines and organic acids, both derived from acyl-CoAs, are excellent markers of most CAMDs. Clinically, each CAMD is unique but strikingly, three main patterns emerge: first, systemic decompensations with combinations of acidosis, ketosis, hypoglycemia, hyperammonemia and fatty liver; second, neurological episodes, particularly acute "stroke-like" episodes, often involving the basal ganglia but sometimes cerebral cortex, brainstem or optic nerves and third, especially in CAMDs of long chain fatty acyl-CoA metabolism, lipid myopathy, cardiomyopathy and arrhythmia. Some patients develop signs from more than one category. The pathophysiology of CAMDs is not precisely understood. Available data suggest that signs may result from CoA sequestration, toxicity and redistribution (CASTOR) in the mitochondrial matrix has been suggested to play a role. This predicts that most CAMDs cause deficiency of CoA, limiting mitochondrial energy production, and that toxic effects from the abnormal accumulation of acyl-CoAs and from extramitochondrial functions of acetyl-CoA may also contribute. Recent progress includes the following. (1) Direct measurements of tissue acyl-CoAs in mammalian models of CAMDs have been related to clinical features. (2) Inborn errors of CoA biosynthesis were shown to cause clinical changes similar to those of inborn errors of acyl-CoA degradation. (3) CoA levels in cells can be influenced pharmacologically. (4) Roles for acetyl-CoA are increasingly identified in all cell compartments. (5) Nonenzymatic acyl-CoA-mediated acylation of intracellular proteins occurs in mammalian tissues and is increased in CAMDs.
Collapse
Affiliation(s)
- Hao Yang
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and Université de Montréal, Canada
| | - Chen Zhao
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and Université de Montréal, Canada; College of Animal Science and Technology, Northwest A&F University, China
| | | | - Youlin Wang
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and Université de Montréal, Canada
| | - Shu Pei Wang
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and Université de Montréal, Canada
| | - Pierre Allard
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and Université de Montréal, Canada
| | | | - Grant A Mitchell
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and Université de Montréal, Canada.
| |
Collapse
|
39
|
OCTN2-Mediated Acetyl-l-Carnitine Transport in Human Pulmonary Epithelial Cells In Vitro. Pharmaceutics 2019; 11:pharmaceutics11080396. [PMID: 31394757 PMCID: PMC6723908 DOI: 10.3390/pharmaceutics11080396] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 12/24/2022] Open
Abstract
The carnitine transporter OCTN2 is associated with asthma and other inflammatory diseases. The aims of this work were (i) to determine carnitine uptake into freshly isolated human alveolar type I (ATI)-like epithelial cells in primary culture, (ii) to compare the kinetics of carnitine uptake between respiratory epithelial in vitro cell models, and (iii) to establish whether any cell line was a suitable model for studies of carnitine transport at the air-blood barrier. Levels of time-dependent [3H]-acetyl-l-carnitine uptake were similar in ATI-like, NCl-H441, and Calu-3 epithelial cells, whereas uptake into A549 cells was ~5 times higher. Uptake inhibition was more pronounced by OCTN2 modulators, such as l-Carnitine and verapamil, in ATI-like primary epithelial cells compared to NCl-H441 and Calu-3 epithelial cells. Our findings suggest that OCTN2 is involved in the cellular uptake of acetyl-l-carnitine at the alveolar epithelium and that none of the tested cell lines are optimal surrogates for primary cells.
Collapse
|
40
|
Hu X, Chandler JD, Park S, Liu K, Fernandes J, Orr M, Smith MR, Ma C, Kang SM, Uppal K, Jones DP, Go YM. Low-dose cadmium disrupts mitochondrial citric acid cycle and lipid metabolism in mouse lung. Free Radic Biol Med 2019; 131:209-217. [PMID: 30529385 PMCID: PMC6331287 DOI: 10.1016/j.freeradbiomed.2018.12.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/30/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022]
Abstract
Cadmium (Cd) causes acute and chronic lung toxicities at occupational exposure levels, yet the impacts of Cd exposure at low levels through dietary intake remain largely uncharacterized. Health concerns arise because humans do not have an effective Cd elimination mechanism, resulting in a long (10- to 35-y) biological half-life. Previous studies showed increased mitochondrial oxidative stress and cell death by Cd yet the details of mitochondrial alterations by low levels of Cd remain unexplored. In the current study, we examined the impacts of Cd burden at a low environmental level on lung metabolome, redox proteome, and inflammation in mice given Cd at low levels by drinking water (0, 0.2, 0.6 and 2.0 mg Cd/L) for 16 weeks. The results showed that mice accumulated lung Cd comparable to non-smoking humans and showed inflammation in lung by histopathology at 2 mg Cd/L. The results of high resolution metabolomics combined with bioinformatics showed that mice treated with 2 mg Cd/L increased levels of lipids in the lung, accompanied by disruption in mitochondrial energy metabolism. In addition, targeted metabolomic analysis showed that these mice had increased accumulation of mitochondrial carnitine and citric acid cycle intermediates. The results of redox proteomics showed that Cd at 2 mg/L stimulated oxidation of isocitrate dehydrogenase, malate dehydrogenase and ATP synthase. Taken together, the results showed impaired mitochondrial function and accumulation of lipids in the lung with a Cd dose response relevant to non-smokers without occupational exposures. These findings suggest that dietary Cd intake could be an important variable contributing to human pulmonary disorders.
Collapse
Affiliation(s)
- Xin Hu
- Division of Pulmonary Medicine, Department of Medicine, Emory University, United States
| | - Joshua D Chandler
- Division of Pulmonary Medicine, Department of Medicine, Emory University, United States
| | - Soojin Park
- Georgia State University, Atlanta, GA, United States
| | - Ken Liu
- Division of Pulmonary Medicine, Department of Medicine, Emory University, United States
| | - Jolyn Fernandes
- Division of Pulmonary Medicine, Department of Medicine, Emory University, United States
| | - Michael Orr
- Division of Pulmonary Medicine, Department of Medicine, Emory University, United States
| | - M Ryan Smith
- Division of Pulmonary Medicine, Department of Medicine, Emory University, United States
| | - Chunyu Ma
- Division of Pulmonary Medicine, Department of Medicine, Emory University, United States
| | - Sang-Moo Kang
- Georgia State University, Atlanta, GA, United States
| | - Karan Uppal
- Division of Pulmonary Medicine, Department of Medicine, Emory University, United States
| | - Dean P Jones
- Division of Pulmonary Medicine, Department of Medicine, Emory University, United States.
| | - Young-Mi Go
- Division of Pulmonary Medicine, Department of Medicine, Emory University, United States.
| |
Collapse
|
41
|
Zhang J, Li Q, Shao Q, Song J, Zhou B, Shu P. Effects of panax notoginseng saponin on the pathological ultrastructure and serum IL‐6 and IL‐8 in pulmonary fibrosis in rabbits. J Cell Biochem 2018; 119:8410-8418. [DOI: 10.1002/jcb.27045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/23/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Jizhou Zhang
- Department of OncologyWenzhou Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medicine UniversityWenzhouZhejiangChina
| | - Qing Li
- Department of Integrated TCM & Western MedicineThe Second Affiliated Hospital of Kunming Medical CollegeKunmingYunnanChina
| | - Qiqi Shao
- Department of NursingWenzhou Lucheng District Wuma Community Health Service CenterWenzhouZhejiangChina
| | - Jingling Song
- Teaching and Research Section of PathologyKunming Medical UniversityKunmingYunnanChina
| | - Bin Zhou
- Department of OncologyWenzhou Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medicine UniversityWenzhouZhejiangChina
| | - Peng Shu
- Department of OncologyJiangsu Province Hospital of TCMNanjingJiangsuChina
| |
Collapse
|
42
|
Showalter MR, Nonnecke EB, Linderholm AL, Cajka T, Sa MR, Lönnerdal B, Kenyon NJ, Fiehn O. Obesogenic diets alter metabolism in mice. PLoS One 2018; 13:e0190632. [PMID: 29324762 PMCID: PMC5764261 DOI: 10.1371/journal.pone.0190632] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/18/2017] [Indexed: 12/22/2022] Open
Abstract
Obesity and accompanying metabolic disease is negatively correlated with lung health yet the exact mechanisms by which obesity affects the lung are not well characterized. Since obesity is associated with lung diseases as chronic bronchitis and asthma, we designed a series of experiments to measure changes in lung metabolism in mice fed obesogenic diets. Mice were fed either control or high fat/sugar diet (45%kcal fat/17%kcal sucrose), or very high fat diet (60%kcal fat/7% sucrose) for 150 days. We performed untargeted metabolomics by GC-TOFMS and HILIC-QTOFMS and lipidomics by RPLC-QTOFMS to reveal global changes in lung metabolism resulting from obesity and diet composition. From a total of 447 detected metabolites, we found 91 metabolite and lipid species significantly altered in mouse lung tissues upon dietary treatments. Significantly altered metabolites included complex lipids, free fatty acids, energy metabolites, amino acids and adenosine and NAD pathway members. While some metabolites were altered in both obese groups compared to control, others were different between obesogenic diet groups. Furthermore, a comparison of changes between lung, kidney and liver tissues indicated few metabolic changes were shared across organs, suggesting the lung is an independent metabolic organ. These results indicate obesity and diet composition have direct mechanistic effects on composition of the lung metabolome, which may contribute to disease progression by lung-specific pathways.
Collapse
Affiliation(s)
- Megan R. Showalter
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, United States of America
| | - Eric B. Nonnecke
- Department of Nutrition, University of California Davis, Davis, CA, United States of America
| | - A. L. Linderholm
- Department of Internal Medicine, Division of Pulmonary and Critical Care, University of California Davis, Davis, CA, United States of America
| | - Tomas Cajka
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, United States of America
| | - Michael R. Sa
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, United States of America
| | - Bo Lönnerdal
- Department of Nutrition, University of California Davis, Davis, CA, United States of America
| | - Nicholas J. Kenyon
- Department of Internal Medicine, Division of Pulmonary and Critical Care, University of California Davis, Davis, CA, United States of America
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, United States of America
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- * E-mail:
| |
Collapse
|
43
|
Shan J, Qian W, Shen C, Lin L, Xie T, Peng L, Xu J, Yang R, Ji J, Zhao X. High-resolution lipidomics reveals dysregulation of lipid metabolism in respiratory syncytial virus pneumonia mice. RSC Adv 2018; 8:29368-29377. [PMID: 35548018 PMCID: PMC9084459 DOI: 10.1039/c8ra05640d] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/02/2018] [Indexed: 11/21/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a leading viral pathogen responsible for lower respiratory tract infections, particularly in children under five years worldwide, often resulting in hospitalization. At present, the molecular-level interactions between RSV and its host and the underlying mechanisms of RSV-induced inflammation are poorly understood. Herein, we describe an untargeted high-resolution lipidomics platform based on UHPLC-Q-Exactive-MS to assess the lipid alterations of lung tissues and plasma from a mouse model of RSV pneumonia. Untargeted lipidomics using LC-MS with multivariate analysis was applied to describe the lipidomic profiling of the lung tissues and plasma in RSV pneumonia mice. Lipid identification was conducted via an in silico MS/MS LipidBlast library using the MS-DIAL software. We observed distinct compartmental lipid signatures in the mice lung tissues and plasma and significant lipid profile changes between the systematic and localized host responses to RSV. A total of 87 and 68 differential lipids were captured in the mice lung tissue and plasma, respectively, including phospholipids, sphingolipids, acylcarnitine, and fatty acids. Some of these lipids belong to pulmonary surfactants, illustrating that RSV pneumonia-induced aberrations of the pulmonary surfactant system may play a vital role in the etiology of respiratory inflammation. Our findings reveal that the host responses to RSV and various lipid metabolic pathways were linked to disease pathology. Furthermore, our findings could provide mechanistic insights into RSV pneumonia. Respiratory syncytial virus (RSV) is a leading viral pathogen responsible for lower respiratory tract infections, particularly in children under five years worldwide, often resulting in hospitalization.![]()
Collapse
|
44
|
Advances in the Understanding and Treatment of Mitochondrial Fatty Acid Oxidation Disorders. CURRENT GENETIC MEDICINE REPORTS 2017; 5:132-142. [PMID: 29177110 DOI: 10.1007/s40142-017-0125-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose of review This review focuses on advances made in the past three years with regards to understanding the mitochondrial fatty acid oxidation (FAO) pathway, the pathophysiological ramifications of genetic lesions in FAO enzymes, and emerging therapies for FAO disorders. Recent findings FAO has now been recognized to play a key energetic role in pulmonary surfactant synthesis, T-cell differentiation and memory, and the response of the proximal tubule to kidney injury. Patients with FAO disorders may face defects in these cellular systems as they age. Aspirin, statins, and nutritional supplements modulate the rate of FAO under normal conditions and could be risk factors for triggering symptoms in patients with FAO disorders. Patients have been identified with mutations in the ACAD9 and ECHS1 genes, which may represent new FAO disorders. New interventions for long-chain FAODs are in clinical trials. Finally, post-translational modifications that regulate fatty acid oxidation protein activities have been characterized that represent important new therapeutic targets. Summary Recent research has led to a deeper understanding of FAO. New therapeutic avenues are being pursued that may ultimately cause a paradigm shift for patient care.
Collapse
|
45
|
Mass spectrometry imaging identifies palmitoylcarnitine as an immunological mediator during Salmonella Typhimurium infection. Sci Rep 2017; 7:2786. [PMID: 28584281 PMCID: PMC5459799 DOI: 10.1038/s41598-017-03100-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/25/2017] [Indexed: 12/19/2022] Open
Abstract
Salmonella Typhimurium causes a self-limiting gastroenteritis that may lead to systemic disease. Bacteria invade the small intestine, crossing the intestinal epithelium from where they are transported to the mesenteric lymph nodes (MLNs) within migrating immune cells. MLNs are an important site at which the innate and adaptive immune responses converge but their architecture and function is severely disrupted during S. Typhimurium infection. To further understand host-pathogen interactions at this site, we used mass spectrometry imaging (MSI) to analyse MLN tissue from a murine model of S. Typhimurium infection. A molecule, identified as palmitoylcarnitine (PalC), was of particular interest due to its high abundance at loci of S. Typhimurium infection and MLN disruption. High levels of PalC localised to sites within the MLNs where B and T cells were absent and where the perimeter of CD169+ sub capsular sinus macrophages was disrupted. MLN cells cultured ex vivo and treated with PalC had reduced CD4+CD25+ T cells and an increased number of B220+CD19+ B cells. The reduction in CD4+CD25+ T cells was likely due to apoptosis driven by increased caspase-3/7 activity. These data indicate that PalC significantly alters the host response in the MLNs, acting as a decisive factor in infection outcome.
Collapse
|
46
|
Brandsma J, Postle AD. Analysis of the regulation of surfactant phosphatidylcholine metabolism using stable isotopes. Ann Anat 2017; 211:176-183. [PMID: 28351529 DOI: 10.1016/j.aanat.2017.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 01/22/2023]
Abstract
The pathways and mechanisms that regulate pulmonary surfactant synthesis, processing, secretion and catabolism have been extensively characterised using classical biochemical and analytical approaches. These have constructed a model, largely in experimental animals, for surfactant phospholipid metabolism in the alveolar epithelial cell whereby phospholipid synthesised on the endoplasmic reticulum is selectively transported to lamellar body storage vesicles, where it is subsequently processed before secretion into the alveolus. Surfactant phospholipid is a complex mixture of individual molecular species defined by the combination of esterified fatty acid groups and a comprehensive description of surfactant phospholipid metabolism requires consideration of the interactions between such molecular species. However, until recently, lipid analytical techniques have not kept pace with the considerable advances in understanding of the enzymology and molecular biology of surfactant metabolism. Refinements in electrospray ionisation mass spectrometry (ESI-MS) can now provide very sensitive platforms for the rapid characterisation of surfactant phospholipid composition in molecular detail. The combination of ESI-MS and administration of phospholipid substrates labelled with stable isotopes extends this analytical approach to the quantification of synthesis and turnover of individual molecular species of surfactant phospholipid. As this methodology does not involve radioactivity, it is ideally suited to application in clinical studies. This review will provide an overview of the metabolic processes that regulate the molecular specificity of surfactant phosphatidylcholine together with examples of how the application of stable isotope technologies in vivo has, for the first time, begun to explore regulation of the molecular specificity of surfactant synthesis in human subjects.
Collapse
Affiliation(s)
- Joost Brandsma
- Academic Unit of Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom
| | - Anthony D Postle
- Academic Unit of Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom.
| |
Collapse
|
47
|
Affiliation(s)
- Claude A. Piantadosi
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina 27710;
| | - Hagir B. Suliman
- Departments of Anesthesiology and Pathology, Duke University School of Medicine, Durham, North Carolina 27710;
| |
Collapse
|
48
|
Kellner M, Noonepalle S, Lu Q, Srivastava A, Zemskov E, Black SM. ROS Signaling in the Pathogenesis of Acute Lung Injury (ALI) and Acute Respiratory Distress Syndrome (ARDS). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:105-137. [PMID: 29047084 PMCID: PMC7120947 DOI: 10.1007/978-3-319-63245-2_8] [Citation(s) in RCA: 272] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The generation of reactive oxygen species (ROS) plays an important role for the maintenance of cellular processes and functions in the body. However, the excessive generation of oxygen radicals under pathological conditions such as acute lung injury (ALI) and its most severe form acute respiratory distress syndrome (ARDS) leads to increased endothelial permeability. Within this hallmark of ALI and ARDS, vascular microvessels lose their junctional integrity and show increased myosin contractions that promote the migration of polymorphonuclear leukocytes (PMNs) and the transition of solutes and fluids in the alveolar lumen. These processes all have a redox component, and this chapter focuses on the role played by ROS during the development of ALI/ARDS. We discuss the origins of ROS within the cell, cellular defense mechanisms against oxidative damage, the role of ROS in the development of endothelial permeability, and potential therapies targeted at oxidative stress.
Collapse
Affiliation(s)
- Manuela Kellner
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA
| | - Satish Noonepalle
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA
| | - Qing Lu
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA
| | - Anup Srivastava
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA
| | - Evgeny Zemskov
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA
| | - Stephen M Black
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA.
| |
Collapse
|
49
|
Abstract
Mitochondria are a distinguishing feature of eukaryotic cells. Best known for their critical function in energy production via oxidative phosphorylation (OXPHOS), mitochondria are essential for nutrient and oxygen sensing and for the regulation of critical cellular processes, including cell death and inflammation. Such diverse functional roles for organelles that were once thought to be simple may be attributed to their distinct heteroplasmic genome, exclusive maternal lineage of inheritance, and ability to generate signals to communicate with other cellular organelles. Mitochondria are now thought of as one of the cell's most sophisticated and dynamic responsive sensing systems. Specific signatures of mitochondrial dysfunction that are associated with disease pathogenesis and/or progression are becoming increasingly important. In particular, the centrality of mitochondria in the pathological processes and clinical phenotypes associated with a range of lung diseases is emerging. Understanding the molecular mechanisms regulating the mitochondrial processes of lung cells will help to better define phenotypes and clinical manifestations associated with respiratory disease and to identify potential diagnostic and therapeutic targets.
Collapse
|
50
|
Dambrova M, Makrecka-Kuka M, Vilskersts R, Makarova E, Kuka J, Liepinsh E. Pharmacological effects of meldonium: Biochemical mechanisms and biomarkers of cardiometabolic activity. Pharmacol Res 2016; 113:771-780. [PMID: 26850121 DOI: 10.1016/j.phrs.2016.01.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/13/2016] [Accepted: 01/15/2016] [Indexed: 01/07/2023]
Abstract
Meldonium (mildronate; 3-(2,2,2-trimethylhydrazinium)propionate; THP; MET-88) is a clinically used cardioprotective drug, which mechanism of action is based on the regulation of energy metabolism pathways through l-carnitine lowering effect. l-Carnitine biosynthesis enzyme γ-butyrobetaine hydroxylase and carnitine/organic cation transporter type 2 (OCTN2) are the main known drug targets of meldonium, and through inhibition of these activities meldonium induces adaptive changes in the cellular energy homeostasis. Since l-carnitine is involved in the metabolism of fatty acids, the decline in its levels stimulates glucose metabolism and decreases concentrations of l-carnitine related metabolites, such as long-chain acylcarnitines and trimethylamine-N-oxide. Here, we briefly reviewed the pharmacological effects and mechanisms of meldonium in treatment of heart failure, myocardial infarction, arrhythmia, atherosclerosis and diabetes.
Collapse
Affiliation(s)
- Maija Dambrova
- Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, Riga LV-1006, Latvia; Riga Stradins University, Dzirciema Str. 16, Riga LV-1007, Latvia.
| | - Marina Makrecka-Kuka
- Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, Riga LV-1006, Latvia
| | - Reinis Vilskersts
- Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, Riga LV-1006, Latvia; Riga Stradins University, Dzirciema Str. 16, Riga LV-1007, Latvia
| | - Elina Makarova
- Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, Riga LV-1006, Latvia
| | - Janis Kuka
- Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, Riga LV-1006, Latvia
| | - Edgars Liepinsh
- Latvian Institute of Organic Synthesis, Aizkraukles Str. 21, Riga LV-1006, Latvia
| |
Collapse
|