51
|
Li N, Huang Y, Chen F, Yin Z, Wang X, Zhang K. Metabolome and microbiome analyses reveal the efficacy of Shen-Fu formula in treating heart failure. Biomed Chromatogr 2024; 38:e6032. [PMID: 39468419 DOI: 10.1002/bmc.6032] [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: 06/13/2024] [Revised: 09/27/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024]
Abstract
Improvement of strategies to treat heart failure (HF) has been a longstanding global goal and challenge. Shen-Fu formula (SF), as a classic herbal preparation, has demonstrated efficacy in treating HF in clinical settings. However, further understanding of the therapeutic mechanisms of SF is required. In this study, metabolomics and 16S rDNA sequencing were used to analyze the effects of SF on metabolic profiling and gut microbiota in HF rats. After 4 weeks of SF treatment, the cardiac function of HF rats showed improvement, with a significant increase in ejection fraction and fractional shortening, as well as a significant decrease in left ventricular volume and mass. Metabolomics study revealed that SF regulates the levels of substances related to energy metabolism, primarily involving lysophosphatidylcholines and polyunsaturated fatty acids. In addition, we found that SF regulates the structure of the microbial community in HF rats and modulates the balance between probiotic and pathogenic bacteria. Furthermore, the SF combination exhibited a superior effect that was better than the use of each herb separately. These results demonstrate the potential of SF therapy in the management of HF and highlight the role of SF in regulating fatty acid metabolism and gut microbiome during HF.
Collapse
Affiliation(s)
- Nana Li
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Yuting Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Feng Chen
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhaorui Yin
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiao Wang
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Kai Zhang
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
52
|
Hsu RH, Lee NC, Chen HA, Hwu WL, Lee WT, Chien YH. Ataluren-mediated nonsense variant readthrough in D-bifunctional protein deficiency: A case report. Mol Genet Metab Rep 2024; 41:101137. [PMID: 39282052 PMCID: PMC11402207 DOI: 10.1016/j.ymgmr.2024.101137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/18/2024] Open
Abstract
D-bifunctional protein (DBP) deficiency, a fatal peroxisomal enzyme disorder, typically manifests with life-threatening symptoms in the first two years of childhood. We present the case of an infant with elevated lysophosphatidylcholine C26:0 (C26:0-LPC) levels identified during X-linked adrenoleukodystrophy (ALD) screening, leading to a diagnosis of DBP deficiency due to a homozygous HSD17B4 c.1041T>A, p.(Tyr347Ter) variant. Starting at two months of age, the infant experienced seizures, hypotonia, and developmental delays, prompting the initiation of experimental treatment with the readthrough agent PTC124 (ataluren) at six months. The treatment led to a decrease in C26:0-LPC levels from 0.65 μM to 0.53 μM; concomitant fish oil supplementation transiently increased C26:0-LPC to 0.74 μM before returning to 0.53 μM after cessation of supplementation. The patient demonstrated improved swallowing and progressive motor and speech development during a two-year treatment period, with no further seizures. This case report highlights the potential of nonsense readthrough therapy for peroxisomal disorders, a group of metabolic diseases that currently lack targeted treatments.
Collapse
Affiliation(s)
- Rai-Hseng Hsu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ni-Chung Lee
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hui-An Chen
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wuh-Liang Hwu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Center for Precision Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wang-Tso Lee
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yin-Hsiu Chien
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
53
|
Fowle-Grider R, Rowles JL, Shen I, Wang Y, Schwaiger-Haber M, Dunham AJ, Jayachandran K, Inkman M, Zahner M, Naser FJ, Jackstadt MM, Spalding JL, Chiang S, McCommis KS, Dolle RE, Kramer ET, Zimmerman SM, Souroullas GP, Finck BN, Shriver LP, Kaufman CK, Schwarz JK, Zhang J, Patti GJ. Dietary fructose enhances tumour growth indirectly via interorgan lipid transfer. Nature 2024; 636:737-744. [PMID: 39633044 DOI: 10.1038/s41586-024-08258-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/21/2024] [Indexed: 12/07/2024]
Abstract
Fructose consumption has increased considerably over the past five decades, largely due to the widespread use of high-fructose corn syrup as a sweetener1. It has been proposed that fructose promotes the growth of some tumours directly by serving as a fuel2,3. Here we show that fructose supplementation enhances tumour growth in animal models of melanoma, breast cancer and cervical cancer without causing weight gain or insulin resistance. The cancer cells themselves were unable to use fructose readily as a nutrient because they did not express ketohexokinase-C (KHK-C). Primary hepatocytes did express KHK-C, resulting in fructolysis and the excretion of a variety of lipid species, including lysophosphatidylcholines (LPCs). In co-culture experiments, hepatocyte-derived LPCs were consumed by cancer cells and used to generate phosphatidylcholines, the major phospholipid of cell membranes. In vivo, supplementation with high-fructose corn syrup increased several LPC species by more than sevenfold in the serum. Administration of LPCs to mice was sufficient to increase tumour growth. Pharmacological inhibition of ketohexokinase had no direct effect on cancer cells, but it decreased circulating LPC levels and prevented fructose-mediated tumour growth in vivo. These findings reveal that fructose supplementation increases circulating nutrients such as LPCs, which can enhance tumour growth through a cell non-autonomous mechanism.
Collapse
Affiliation(s)
- Ronald Fowle-Grider
- Department of Chemistry, Washington University, St Louis, MO, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Joe L Rowles
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Isabel Shen
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Yahui Wang
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Michaela Schwaiger-Haber
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Alden J Dunham
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Kay Jayachandran
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Matthew Inkman
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Michael Zahner
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Fuad J Naser
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Madelyn M Jackstadt
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Jonathan L Spalding
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Sarah Chiang
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Kyle S McCommis
- Department of Biochemistry & Molecular Biology, Saint Louis University School of Medicine, St Louis, MO, USA
| | - Roland E Dolle
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Eva T Kramer
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Sarah M Zimmerman
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - George P Souroullas
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
| | - Brian N Finck
- Division of Geriatrics and Nutritional Sciences, Washington University School of Medicine, St Louis, MO, USA
| | - Leah P Shriver
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Charles K Kaufman
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Julie K Schwarz
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jin Zhang
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
- Institute for Informatics, Data Science & Biostatistics (I2DB), Washington University School of Medicine, St Louis, MO, USA
| | - Gary J Patti
- Department of Chemistry, Washington University, St Louis, MO, USA.
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA.
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA.
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
54
|
Ren J, Wang X, Sun Y, Yang L, Sun H, Sun Y, Kong L, Yan G, Han Y, Wang X. Integrated metabolomics and lipidomics investigation of the mechanism of Danggui Sini Decoction on improving lipid homeostasis in primary dysmenorrhea. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156034. [PMID: 39306882 DOI: 10.1016/j.phymed.2024.156034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/26/2024] [Accepted: 09/07/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Danggui Sini Decoction (DGSND) is a classic prescription for treating primary dysmenorrhea (PD), while, the ameliorating effects of DGSND on PD and its mechanisms are not yet fully understood. PURPOSE The present study is devoted to investigate the protective effect of DGSND against PD and the possible mechanism from the perspective of metabolomics as well as lipidomics. METHODS DGSND was characterized by UPLC-Q-TOF/MS. The PD rat model was induced by estradiol benzoate and oxytocin, and traditional pharmacology, including writhing times, latency time, biochemical index, organ index, and histopathology were performed to evaluated the efficacy of DGSND on PD. Urine metabolomics strategy combined with functional analysis was adopted to delineate the therapeutic effect of DGSND on PD rats and anchor the crucial pathway, and lipidomics analysis was further performed with the uterine tissue as the research object to elucidate the protective mechanism of DGSND from the perspective of lipid homeostasis. Finally, western blot analysis was used to validate the expression of key metabolic enzymes in lipid metabolism. RESULTS DGSND was effective in ameliorating writhing times, latency time, the value of prostaglandin F2α (PGF2α)/PGE2, uterus index, and morphological changes of PD rats. Metabolic signature of PD rats was primarily characterized by the disturbance of steroid hormone metabolism, amino acid metabolism, and lipid metabolism. Functional analysis revealed the urine biomarkers of PD were most related with lipid abnormality. Further lipidomics analysis indicated DGSND exerted anti-PD effects by remodeling lipid homeostasis, which might be due to the significant correlations between different kinds of lipids, especially the extremely high correlation of phosphatidylethanolamine, phosphatidylcholine, and fatty acids. Moreover, the key metabolic enzymes expression of CK, PLA2, LPCAT3, COX-2, and 5-LOX can be greatly downregulated by DGSND. CONCLUSION Our findings demonstrated a novel protective mechanism of DGSND against PD by regulating lipid homeostasis.
Collapse
Affiliation(s)
- Junling Ren
- State key laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, PR China
| | - Xia Wang
- State key laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, PR China
| | - Yuran Sun
- State key laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, PR China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, PR China
| | - Hui Sun
- State key laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, PR China.
| | - Ye Sun
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, PR China
| | - Ling Kong
- State key laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, PR China
| | - Guangli Yan
- State key laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, PR China
| | - Ying Han
- State key laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, PR China
| | - Xijun Wang
- State key laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, PR China; State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, PR China.
| |
Collapse
|
55
|
Perry AS, Piaggi P, Huang S, Nayor M, Freedman J, North KE, Below JE, Clish CB, Murthy VL, Krakoff J, Shah RV. Human metabolic chambers reveal a coordinated metabolic-physiologic response to nutrition. JCI Insight 2024; 9:e184279. [PMID: 39576013 PMCID: PMC11601946 DOI: 10.1172/jci.insight.184279] [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: 06/27/2024] [Accepted: 09/25/2024] [Indexed: 11/27/2024] Open
Abstract
Human studies linking metabolism with organism-wide physiologic function have been challenged by confounding, adherence, and precisionHere, we united physiologic and molecular phenotypes of metabolism during controlled dietary intervention to understand integrated metabolic-physiologic responses to nutrition. In an inpatient study of individuals who underwent serial 24-hour metabolic chamber experiments (indirect calorimetry) and metabolite profiling, we mapped a human metabolome onto substrate oxidation rates and energy expenditure across up to 7 dietary conditions (energy balance, fasting, multiple 200% caloric excess overfeeding of varying fat, protein, and carbohydrate composition). Diets exhibiting greater fat oxidation (e.g., fasting, high-fat) were associated with changes in metabolites within pathways of mitochondrial β-oxidation, ketogenesis, adipose tissue fatty acid liberation, and/or multiple anapleurotic substrates for tricarboxylic acid cycle flux, with inverse associations for diets with greater carbohydrate availability. Changes in each of these metabolite classes were strongly related to 24-hour respiratory quotient (RQ) and substrate oxidation rates (e.g., acylcarnitines related to lower 24-hour RQ and higher 24-hour lipid oxidation), underscoring links between substrate availability, physiology, and metabolism in humans. Physiologic responses to diet determined by gold-standard human metabolic chambers are strongly coordinated with biologically consistent, interconnected metabolic pathways encoded in the metabolome.
Collapse
Affiliation(s)
- Andrew S. Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona, USA
| | - Shi Huang
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jane Freedman
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kari E. North
- CVD Genetic Epidemiology Computational Laboratory, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jennifer E. Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Clary B. Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts USA
| | | | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona, USA
| | - Ravi V. Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
56
|
Ortlund E, Hou Z, Chen CY, Gaul D, Zhang T, Moore S, Liu X, Ivanova A, Maner-Smith K, Newgard C, Bodine S, Savage E, Bennett A, Fernandez F. Endurance Exercise Training Alters Lipidomic Profiles of Plasma and Eight Tissues in Rats: a MoTrPAC study. RESEARCH SQUARE 2024:rs.3.rs-5263273. [PMID: 39606465 PMCID: PMC11601870 DOI: 10.21203/rs.3.rs-5263273/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Endurance exercise training (ExT) induces metabolic, structural, and functional adaptations via lipidomic modifications, yet the systematic elucidation of lipidome alterations in response to ExT remains incomplete. As a part of the Molecular Transducers of Physical Activity Consortium (MoTrPAC), we leveraged non-targeted and targeted lipidomics for the systematic discovery of lipid alterations in the brown adipose tissue, heart, hippocampus, kidney, liver, lung, skeletal muscle gastrocnemius, subcutaneous white adipose tissue, and plasma in response to 1, 2, 4 or 8 weeks of ExT in 6-month-old male and female Fischer-344 rats. This study demonstrates that these tissues, each with distinct lipidomic features, underwent dynamic, sexually dimorphic lipid remodeling. Exercise trained animals showed reduced whole-body adiposity and improved cardiorespiratory fitness, along with enhanced utilization of lipid stores and dynamic triacylglycerol remodeling compared to sedentary controls in all tissues except hippocampus. They also showed modifications in phospholipids, lysophospholipids, oxylipins, and ceramides in several tissues. Coordinated changes across tissues reflect systemic tissue communication, with liver-plasma-heart connection potentially playing a key role in systemic lipid metabolism during ExT. These data will improve our understanding of lipid-associated biological processes underlying the health-promoting benefits of ExT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anna Ivanova
- Centers for Disease Control and Prevention (CDC)
| | | | | | | | | | | | | |
Collapse
|
57
|
Wang S, Liu C, Ding R, Wang S, Ye Y, He M. Alterations in Gut Microbiota and Serum Metabolites in Children with Mycoplasma pneumoniae Pneumonia. Infect Drug Resist 2024; 17:5097-5110. [PMID: 39584178 PMCID: PMC11585984 DOI: 10.2147/idr.s490547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024] Open
Abstract
Background Over the past years, there has been a significant increase in the incidence of Mycoplasma pneumoniae (MP) infections, particularly among pediatric patients, nationwide. An emerging body of research has established a link between dysbiosis of the host microbiome and the metabolic functioning of the host, which contributes to the development of respiratory diseases. Methods A total of 25 children were included in the study, comprising 15 pneumonia patients and 10 healthy children. Stool samples were collected from all participants to analyze the 16S ribosomal RNA (16S rRNA) gene, while serum samples were prepared for untargeted metabolomics to qualitatively and quantitatively assess short-chain fatty acids. Results The gut microbial composition of individuals with Mycoplasma pneumoniae pneumonia (MPP) exhibited significant differences compared to healthy children. Notably, diseased children demonstrated higher microbial diversity and an enrichment of opportunistic pathogens, such as Erysipelatoclostridium and Eggerthella. Analysis revealed elevated levels of two specific short-chain fatty acids, namely acetic acid and isobutyric acid, in the MPP group, suggesting their potential as biomarkers for predicting MP infection. Metabolomic signature analysis identified a significant increase in major classes of glycerophospholipids in the MPP group. Moreover, we identified a total of 750 significant correlations between gut microbiota and circulating serum metabolites. MPP enriched genera Erysipelatoclostridium and Eggerthella, exhibited negative associations with indole-3-butyric acid. Additionally, Eggerthella showed a positive correlation with inflammatory metabolites LPC (18:0). Discussion Collectively, these findings provide novel insights into the selection of potential biomarkers and the pathogenesis of MPP in children based on the gut microbiota and systemic circulating metabolites.
Collapse
Affiliation(s)
- Shu Wang
- Department of Geriatrics, The First People’s Hospital of Hefei, Hefei, 230061, People’s Republic of China
| | - Chengzhong Liu
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Ruipei Ding
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Shumei Wang
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Yousheng Ye
- Department of Geriatrics, The First People’s Hospital of Hefei, Hefei, 230061, People’s Republic of China
| | - Maozhang He
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, People’s Republic of China
| |
Collapse
|
58
|
Longo S, Cicalini I, Pieragostino D, De Laurenzi V, Legramante JM, Menghini R, Rizza S, Federici M. A Metabolomic Approach to Unexplained Syncope. Biomedicines 2024; 12:2641. [PMID: 39595205 PMCID: PMC11591916 DOI: 10.3390/biomedicines12112641] [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: 10/03/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Background: This study aims to identify a metabolomic signature that facilitates the classification of syncope and the categorization of the unexplained syncope (US) to aid in its management. Methods: We compared a control group (CTRL, n = 10) with a transient loss of consciousness (TLC) group divided into the OH group (n = 23) for orthostatic syncope, the NMS group (n = 26) for neuromediated syncope, the CS group (n = 9) for cardiological syncope, and the US group (n = 27) for US defined as syncope without a precise categorization after first- and second-level diagnostic approaches. Results: The CTRL and the TLC groups significantly differed in metabolic profile. A new logistic regression model has been developed to predict how the US will be clustered. Using differences in lysophosphatidylcholine with 22 carbon atom (C22:0-LPC) levels, 96% of the US belongs to the NMS and 4% to the CS subgroup. Differences in glutamine and lysine (GLN/LYS) levels clustered 95% of the US in the NMS and 5% in the CS subgroup. Conclusions: We hypothesize a possible role of C22:0 LPC and GLN/LYS in re-classifying US and differentiating it from cardiological syncope.
Collapse
Affiliation(s)
- Susanna Longo
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (S.L.); (J.M.L.); (R.M.); (S.R.)
| | - Ilaria Cicalini
- Department of Innovative Technologies in Medicine and Dentistry, “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (D.P.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Damiana Pieragostino
- Department of Innovative Technologies in Medicine and Dentistry, “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (D.P.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine and Dentistry, “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (D.P.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Jacopo M. Legramante
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (S.L.); (J.M.L.); (R.M.); (S.R.)
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (S.L.); (J.M.L.); (R.M.); (S.R.)
| | - Stefano Rizza
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (S.L.); (J.M.L.); (R.M.); (S.R.)
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (S.L.); (J.M.L.); (R.M.); (S.R.)
| |
Collapse
|
59
|
Estevao IL, Kazman JB, Bramer LM, Nicora C, Ren MQ, Sambuughin N, Munoz N, Kim YM, Bloodsworth K, Richert M, Teeguarden J, Burnum-Johnson K, Deuster PA, Nakayasu ES, Many G. The human plasma lipidome response to exertional heat tolerance testing. Lipids Health Dis 2024; 23:380. [PMID: 39548465 PMCID: PMC11566608 DOI: 10.1186/s12944-024-02322-7] [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: 06/07/2024] [Accepted: 10/01/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND The year of 2023 displayed the highest average global temperatures since it has been recorded-the duration and severity of extreme heat are projected to increase. Rising global temperatures represent a major public health threat, especially to occupations exposed to hot environments, such as construction and agricultural workers, and first responders. Despite efforts of the scientific community, there is still a need to characterize the pathophysiological processes leading to heat related illness and develop biomarkers that can predict its onset. METHODS Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based lipidomics analysis was performed on plasma from male and female subjects who underwent exertional heat tolerance testing (HTT), consisting of a 2-h treadmill walk at 5 km/h with 2.0% incline at a controlled temperature of 40ºC. From HTT, heat tolerance was calculated using the physiological strain index (PSI). RESULTS Nearly half of all 995 detected lipids from 27 classes were responsive to HTT. Lipid classes related to substrate utilization were predominantly affected by HTT, with a downregulation of triacylglycerols and upregulation of free fatty acids and acyl-carnitines (CARs). Even chain CAR 4:0, 14:0 and 16:1, suggested by-products of incomplete beta oxidation, and diacylglycerols displayed the highest correlation to PSI. PSI did not correlate with plasma lactate levels, suggesting that correlations between even chain CARs and PSI are related to metabolic efficiency versus physical exertion. CONCLUSIONS Overall, HTT displays a strong impact on the human plasma lipidome and lipid metabolic inefficiencies may underlie reduced heat tolerance.
Collapse
Affiliation(s)
| | - Josh B Kazman
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Lisa M Bramer
- Biological Sciences Division, Richland, WA, 99352, USA
| | - Carrie Nicora
- Biological Sciences Division, Richland, WA, 99352, USA
| | - Ming Qiang Ren
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Nyamkhishig Sambuughin
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Nathalie Munoz
- Environmental and Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Young-Mo Kim
- Biological Sciences Division, Richland, WA, 99352, USA
| | | | - Maile Richert
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Justin Teeguarden
- Environmental and Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Kristin Burnum-Johnson
- Environmental and Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Patricia A Deuster
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | | | - Gina Many
- Biological Sciences Division, Richland, WA, 99352, USA.
| |
Collapse
|
60
|
Wu Q, Liu K, Hou R, Wu X, Ruan X, Wang M, Sun Z, Meng L, Dai G, Li C, Wu J, Mu G. Integrative Lipid Pseudotargeted Metabolomics and Amino Acids Targeted Metabolomics Unravel the Therapeutic Mechanism of Rhizoma Paridis Saponins on Experimental Colitis of Damp-Heat Type. Drug Des Devel Ther 2024; 18:5087-5108. [PMID: 39554759 PMCID: PMC11568858 DOI: 10.2147/dddt.s476494] [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: 07/19/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose Inflammatory bowel disease (IBD) is a serious disease that affects the metabolism and inflammatory responses of human beings. From the perspective of traditional Chinese medicine, damp-heat syndrome is one of the main syndromes of IBD. Rhizoma Paridis, also known as the root of Paris polyphylla, a well-known herbal medicine used in China, is used to treat IBD with damp-heat syndrome (IBD-DH). However, uncertainty still exists regarding the underlying mechanisms and the impact of Rhizoma Paridis on IBD-DH. Methods The rats in the model (DAT) and medication administration (Rhizoma Paridis total saponins (RPTS) and Pennogenin (PN)) groups were given a high temperature and high humidity environment, high fat and high sugar diet combined with 2,4,6-trinitrobenzene sulfonic acid (TNBS) to establish the model of experimental colitis of damp-heat type, and the normal control group (RNC) rats were given a normal diet at normal temperature and humidity. Damp-heat control group (DNC) was set with the same condition as DAT without TNBS. Hematoxylin-Eosin (HE) staining was used to observe the histopathological morphology of the rat colorectum. The expression of the metabolism-related genes (Phospholipase A2 (sPLA2, cPLA2), and phosphatidylethanolamine N-methyltransferase (PEMT)) was assessed by using real-time quantitative PCR analysis (RT-qPCR). And the levels of the metabolism-related proteins (sPLA2, cPLA2), S100A8/9, Arg-1, and cytokines were detected by enzyme-linked immunosorbent assay (ELISA) kit. To investigate lipids and amino acids which closely associated with the IBD and IBD-DH, lipid pseudotargeted metabolomics with UHPLC-TQ/MS analysis method, as well as targeted quantitative amino acid analysis were performed. Results Our data showed that RPTS (50 mg/kg) and PN (20 mg/kg) significantly ameliorated the severity of TNBS-induced colitis and downregulated the levels of circulating proinflammatory cytokines. Compared with RNC group, lipid pseudotargeted metabolomics demonstrated that glycerophospholipids, sphingolipids, carnitine, and glycerolipids were the four most perturbed lipid classes, and amino acids targeted metabolomics demonstrated that serine, N-acetylneuraminic acid, histidine, proline, taurine, and kynurenine changed significantly in DAT group . Correlation analyses showed tight associations between most of differential metabolites and proinflammatory cytokines. RPTS and PN both regulated glycerophospholipid metabolism and sphingolipid metabolism. However, both of them did not have a significant effect on amino acid modulation. RPTS and PN potentially regulated sPLA2, cPLA2, and PEMT. Conclusion These results showed that RPTS (50 mg/kg) and PN (20 mg/kg) effectively alleviated rats' colitis of damp-heat type, affected cytokines, and altered lipid metabolism without significant modulation on amino acid metabolism.
Collapse
Affiliation(s)
- Qi Wu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Kexin Liu
- Department of Gastroenterology, The 981th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Chengde, Hebei, 067000, People’s Republic of China
| | - Ruijuan Hou
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Xingxing Wu
- Department of Gastroenterology, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, 450053, People’s Republic of China
| | - Xiaoyu Ruan
- Internal Medicine Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Mao Wang
- Ethics Committee, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Zhiting Sun
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Lingchang Meng
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Guoliang Dai
- Clinical Pharmacology Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Changyin Li
- Clinical Pharmacology Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Jing Wu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Genglin Mu
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| |
Collapse
|
61
|
Feng L, He B, Xia J, Wang Z. Untargeted and Targeted Lipidomics Unveil Dynamic Lipid Metabolism Alterations in Type 2 Diabetes. Metabolites 2024; 14:610. [PMID: 39590846 PMCID: PMC11596168 DOI: 10.3390/metabo14110610] [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: 09/23/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a complex metabolic disorder with a growing body of evidence suggesting the central role of lipid metabolism in its pathogenesis. However, the dynamic changes in lipid metabolism across different stages of T2DM remain understudied. OBJECTIVE This study aimed to elucidate the temporal alterations in lipid metabolism in T2DM using an integrated lipidomics approach. METHOD Serum samples from 155 subjects were analyzed using LC-MS-based lipidomics, including untargeted and targeted approaches. RESULTS We identified significant alterations in 44 lipid metabolites in newly diagnosed T2DM patients and 29 in high-risk individuals, compared with healthy controls. Key metabolic pathways such as sphingomyelin, phosphatidylcholine, and sterol ester metabolism were disrupted, highlighting the involvement of insulin resistance and oxidative stress in T2DM progression. Moreover, 13 lipid metabolites exhibited diagnostic potential for T2DN, showing consistent trends of increase or decrease as the disease progressed. CONCLUSION Our findings underscore the importance of lipid metabolism in T2D development and identify potential lipid biomarkers for early diagnosis and monitoring of disease progression, which contribute to paving the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Li Feng
- School of Agroforestry and Medicine, The Open University of China, Beijing 100039, China;
| | - Bingshu He
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China;
| | - Jianzhen Xia
- School Hospital, Minzu University of China, Beijing 100081, China;
| | - Zhonghua Wang
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China;
| |
Collapse
|
62
|
Guo X, Liu S, Hu W, Lyu X, Xu H, Zhu H, Pan H, Wang L, Wan Y, Yang H, Gong F. The association between metabolite profiles and impaired bone microstructure in adult growth hormone deficient rats. BMC Musculoskelet Disord 2024; 25:883. [PMID: 39508246 PMCID: PMC11539809 DOI: 10.1186/s12891-024-08010-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Adult growth hormone deficiency (AGHD) is associated with an increased risk of fractures and impaired bone microstructure. Understanding the metabolic changes accompanying bone deterioration in AGHD might provide insights into mechanisms behind molecular changes and develop new biomarkers or nutritional strategies for bone destruction. Our study aimed to investigate the association between altered metabolite patterns and impaired bone microstructure in adult rats with growth hormone deficiency. METHODS Thirty seven-week-aged adult Lewis dwarf homozygous (dw/dw) rats (five females and five males), and adult Lewis dwarf heterozygous (dw/ +) rats (five females and five males) rats were compared. Micro-computed tomography (Micro-CT) was used to examine the bone's microstructure. Hematoxylin and eosin (H&E) staining were used to quantify the histological characteristics. Liquid chromatography-mass spectrometry untargeted serum metabolomic analysis was applied in the study. ELISA was used to measure serum bone turnover markers and IGF-1 levels. RESULTS Adult dw/dw rats exhibited great reductions in trabecular volume bone density (Tb.vBMD), bone volume/total volume (BV/TV), and cortical thickness (Ct. Th) compared with adult dw/ + rats (all p values < 0.05), indicating significant impairment in bone microstructure. The serum metabolite profiles revealed substantial differences between the dw/dw rats and dw/ + rats. A total of 134 differential metabolites in positive ion mode and 49 differential metabolites in negative mode were identified. Five metabolites, including Lysophosphatidylcholine(LPC) 20:3, LPC22:6, LPC22:4, cortisol and histamine levels were upregulated in dw/dw rats. The steroid hormone biosynthesis and bile secretion pathways were the main perturbed metabolic pathways. There were significant associations between differential metabolites and the impaired bone microstructure parameters, indicating that the selected metabolites might serve as potential biomarkers for deteriorated bone microstructure in AGHD. CONCLUSION Adult dw/dw rats exhibit impaired bone microstructure and distinct serum metabolic profiles, and the altered metabolites were significantly associated with bone microstructure destruction. This provides a new insight into understanding the mechanism of bone deterioration in AGHD patients from a metabolic perspective.
Collapse
Affiliation(s)
- Xiaonan Guo
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1# Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Shanshan Liu
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1# Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Wenjing Hu
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1# Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Xiaorui Lyu
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1# Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Hanyuan Xu
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1# Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Huijuan Zhu
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1# Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Hui Pan
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1# Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Linjie Wang
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1# Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Yu Wan
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Hongbo Yang
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1# Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| | - Fengying Gong
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1# Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| |
Collapse
|
63
|
Toft-Bertelsen TL, Andreassen SN, Simonsen AH, Hasselbalch SG, MacAulay N. The CSF lipid profile in patients with probable idiopathic normal pressure hydrocephalus differs from control but does not differ between shunt responders and non-responders. Brain Commun 2024; 6:fcae388. [PMID: 39544703 PMCID: PMC11562123 DOI: 10.1093/braincomms/fcae388] [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: 04/19/2024] [Revised: 10/02/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
Idiopathic normal pressure hydrocephalus is a common form of hydrocephalus in the elderly, characterized by enlarged ventricles combined with clinical symptoms presenting as gait impairment, urinary incontinence, and dementia. Idiopathic normal pressure hydrocephalus may be difficult to differentiate clinically from other neurodegenerative disorders, and up to 80% of cases may remain unrecognized and thus untreated. Consequently, there is a pressing demand for biomarkers that can confirm the diagnosis of idiopathic normal pressure hydrocephalus. In this exploratory study, CSF was sampled from the lumbar compartment of 21 control individuals and 19 probable idiopathic normal pressure hydrocephalus patients and analyzed by an untargeted mass spectroscopy-based platform to reveal a complete CSF lipid profile in these samples. Two hundred forty-four lipids from 17 lipid classes were detected in CSF. Various lipid classes, and select individual lipids, were reduced in the CSF obtained from patients with probable idiopathic normal pressure hydrocephalus, whereas a range of lipids belonging to the class of triacylglycerols was elevated. We detected no difference in the CSF lipid profile between probable idiopathic normal pressure hydrocephalus patients with and without clinical improvement following CSF shunting. In conclusion, the lipidomic profile of the CSF in patients with probable idiopathic normal pressure hydrocephalus, therefore, may serve as a sought after biomarker of the pathology, which may be employed to complement the clinical diagnosis.
Collapse
Affiliation(s)
| | | | - Anja Hviid Simonsen
- Department of Neurology, Section 6911, Danish Dementia Research Centre, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
| | - Steen Gregers Hasselbalch
- Department of Neurology, Section 6911, Danish Dementia Research Centre, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
64
|
Zhang G, Zhong X, Chen J, Yang C, Liu Y, Li R, Xu B, Yuan H. The gut microbiome and serum metabolome are altered and interrelated in patients with intracranial atherosclerotic stenosis. J Stroke Cerebrovasc Dis 2024; 33:107887. [PMID: 39208915 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107887] [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: 11/20/2023] [Revised: 07/03/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVES To evaluate the relationship among the gut microbiome, serum metabolites and the Intracranial atherosclerosis stenosis. MATERIALS AND METHODS Integrated analysis of 16S rDNA sequencing of fecal samples and untargeted serum metabolomics was applied to identify alterations in the gut microbiome and serum metabolome in 29 Intracranial atherosclerosis stenosis patients and 29 healthy control individuals. RESULTS Compared to healthy control individuals, the abundances of forty-five genera and one hundred seventy-seven metabolites were significantly altered in Intracranial atherosclerosis stenosis patients. At the species level, the Intracranial atherosclerosis stenosis group exhibited higher abundances of Bacteroidetes and lower abundances of Megaphaera and Muribacoccaceae. Microbial functional prediction analysis revealed enhanced activity of bacterial chemotaxis and oxidative phosphorylation within the Intracranial atherosclerosis stenosis group. In terms of metabolomic findings, the levels of dulcitol were significantly increased in the Intracranial atherosclerosis stenosis group. The levels of specific metabolites within the phosphatidylcholine and lysophosphatidylcholine families, such as PC (14:0e/24:4) and LPC 20:5, were increased, while the levels of certain other specific metabolites were decreased. Dysregulation of certain pathways, such as unsaturated fatty acid metabolism, arginine and proline metabolism may be involved in the development of Intracranial atherosclerosis stenosis. Correlation analysis of the gut microbiome and metabolites revealed a positive correlation between Bacteroides and multiple metabolites, such as Acar 12:3 and PC (8:0/22:6). CONCLUSIONS Our analysis revealed that Bacteroides is a key bacterial genus in gut dysbiosis and may be related to the development of Intracranial atherosclerosis stenosis.
Collapse
Affiliation(s)
- Guangyu Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China
| | - Xiaoling Zhong
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group)
| | - Jing Chen
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group).
| | - Chenli Yang
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group)
| | - Yingbei Liu
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group)
| | - Ran Li
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group)
| | - Bo Xu
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group).
| | - Haicheng Yuan
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group).
| |
Collapse
|
65
|
Lee CW, Tsai HI, Hsu HM, Yu MC, Lee WC, Wang CC, Hsieh YC, Lin CY, Cheng ML, Lo CJ, Wang CT, Lin YC. Do low skeletal muscle bulk and disturbed body fat mass impact tumor recurrence in stage I/II hepatocellular carcinoma undergoing surgery? An observational cohort study. Int J Surg 2024; 110:7067-7079. [PMID: 38959093 PMCID: PMC11573101 DOI: 10.1097/js9.0000000000001905] [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: 05/01/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION The influence of deranged body composition on stage I/II hepatocellular carcinoma (HCC) after surgery remains undetermined. The current study aimed to investigate the impact of low skeletal muscle bulk and disturbed body fat mass on the recurrence outcome of stage I/II HCC patients undergoing liver resection. The associated metabolomic alterations were also assessed. METHODS From 2012 to 2021, stage I and II HCC patients who underwent liver resection at our institute were retrospectively reviewed. Their preoperative body composition including skeletal muscle mass and body fat volume was measured by computed tomography (CT). The recurrence outcome was recorded and analyzed. The preoperative serum was collected and subjected to metabolomic analysis. RESULTS A total of 450 stage I and II HCC patients were included in the current study. Among them, 76% were male and around 60% had HBV infection. After stratified by normal cut-off values obtained from a healthy cohort, 6.4% of stage I/II HCC patients were found to have a low psoas muscle index (PMI), 17.8% a high subcutaneous adipose tissue (SAT) index, and 27.8% a high visceral adipose tissue (VAT) index. Cox regression multivariate analysis further demonstrated that low PMI and high SAT index were independent prognostic factors for time-to-recurrence (TTR) after surgery. Metabolomic analysis discovered that free fatty acid β-oxidation was enhanced in patients with low PMI or high SAT index. CONCLUSION The current study demonstrated that reduced psoas muscle mass may impair while elevated SAT may prolong the TTR of stage I/II HCC patients undergoing liver resections. VAT, on the other hand, was not associated with recurrence outcomes after surgery. Further studies are warranted to validate our findings.
Collapse
Affiliation(s)
- Chao-Wei Lee
- Division of General Surgery, Department of Surgery, Linkou Chang Gung Memorial Hospital
- College of Medicine, Chang Gung University
- Graduate Institute of Clinical Medical Sciences, Chang Gung University
| | - Hsin-I Tsai
- College of Medicine, Chang Gung University
- Graduate Institute of Clinical Medical Sciences, Chang Gung University
- Department of Anesthesiology, Linkou Chang Gung Memorial Hospital
| | - Hsiao-Mei Hsu
- Department of Family Medicine, Taipei City Hospital Zhongxiao Branch, Taipei
| | - Ming-Chin Yu
- College of Medicine, Chang Gung University
- Graduate Institute of Clinical Medical Sciences, Chang Gung University
- Division of General Surgery, Department of Surgery, New Taipei Municipal Tu-Cheng Hospital (Built and Operated by Chang Gung Medical Foundation), Tu-Cheng, New Taipei City
| | - Wei-Chen Lee
- Division of General Surgery, Department of Surgery, Linkou Chang Gung Memorial Hospital
- College of Medicine, Chang Gung University
| | - Chih-Chi Wang
- Division of General Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung
- Division of General Surgery, Department of Surgery, Chiayi Chang Gung Memorial Hospital, Chiayi
| | - Yi-Chung Hsieh
- College of Medicine, Chang Gung University
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital
| | - Cheng-Yu Lin
- College of Medicine, Chang Gung University
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital
| | - Mei-Ling Cheng
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University
- Clinical Metabolomics Core Laboratory, Linkou Chang Gung Memorial Hospital
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Chi-Jen Lo
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University
- Clinical Metabolomics Core Laboratory, Linkou Chang Gung Memorial Hospital
| | - Ching-Ting Wang
- College of Medicine, Chang Gung University
- Department of Nursing, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan
| | - Yu-Ching Lin
- College of Medicine, Chang Gung University
- Department of Medical Imaging and Intervention, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| |
Collapse
|
66
|
Esplin ED, Hanson C, Wu S, Horning AM, Barapour N, Nevins SA, Jiang L, Contrepois K, Lee H, Guha TK, Hu Z, Laquindanum R, Mills MA, Chaib H, Chiu R, Jian R, Chan J, Ellenberger M, Becker WR, Bahmani B, Khan A, Michael B, Weimer AK, Esplin DG, Shen J, Lancaster S, Monte E, Karathanos TV, Ladabaum U, Longacre TA, Kundaje A, Curtis C, Greenleaf WJ, Ford JM, Snyder MP. Multiomic analysis of familial adenomatous polyposis reveals molecular pathways associated with early tumorigenesis. NATURE CANCER 2024; 5:1737-1753. [PMID: 39478120 PMCID: PMC11584401 DOI: 10.1038/s43018-024-00831-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 08/29/2024] [Indexed: 11/24/2024]
Abstract
Familial adenomatous polyposis (FAP) is a genetic disease causing hundreds of premalignant polyps in affected persons and is an ideal model to study transitions of early precancer states to colorectal cancer (CRC). We performed deep multiomic profiling of 93 samples, including normal mucosa, benign polyps and dysplastic polyps, from six persons with FAP. Transcriptomic, proteomic, metabolomic and lipidomic analyses revealed a dynamic choreography of thousands of molecular and cellular events that occur during precancerous transitions toward cancer formation. These involve processes such as cell proliferation, immune response, metabolic alterations (including amino acids and lipids), hormones and extracellular matrix proteins. Interestingly, activation of the arachidonic acid pathway was found to occur early in hyperplasia; this pathway is targeted by aspirin and other nonsteroidal anti-inflammatory drugs, a preventative treatment under investigation in persons with FAP. Overall, our results reveal key genomic, cellular and molecular events during the earliest steps in CRC formation and potential mechanisms of pharmaceutical prophylaxis.
Collapse
Affiliation(s)
- Edward D Esplin
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Casey Hanson
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Si Wu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Aaron M Horning
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Nasim Barapour
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Lihua Jiang
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Hayan Lee
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Tuhin K Guha
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Zheng Hu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | | | - Meredith A Mills
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Hassan Chaib
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Roxanne Chiu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Joanne Chan
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Winston R Becker
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Bahareh Bahmani
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Aziz Khan
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, USA
| | - Basil Michael
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Annika K Weimer
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jeanne Shen
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Samuel Lancaster
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Emma Monte
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Uri Ladabaum
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Teri A Longacre
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Christina Curtis
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - James M Ford
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA.
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA.
| | - Michael P Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
67
|
Wen Y, Li Y, Liu T, Huang L, Yao L, Deng D, Luo W, Cai W, Zhong S, Jin T, Yang X, Wang Q, Wang W, Xue J, Mukherjee R, Hong J, Phillips AR, Windsor JA, Sutton R, Li F, Sun X, Huang W, Xia Q. Chaiqin chengqi decoction treatment mitigates hypertriglyceridemia-associated acute pancreatitis by modulating liver-mediated glycerophospholipid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155968. [PMID: 39217651 DOI: 10.1016/j.phymed.2024.155968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 06/25/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The incidence of hypertriglyceridemia-associated acute pancreatitis (HTG-AP) is increasing globally and more so in China. The characteristics of liver-mediated metabolites and related key enzymes are rarely reported in HTG-AP. Chaiqin chengqi decoction (CQCQD) has been shown to protect against AP including HTG-AP in both patients and rodent models, but the underlying mechanisms in HTG-AP remain unexplored. PURPOSE To assess the characteristics of liver-mediated metabolism and the therapeutic mechanisms of CQCQD in HTG-AP. METHODS Male human apolipoprotein C3 transgenic (hApoC3-Tg; leading to HTG) mice or wild-type littermates received 7 intraperitoneal injections of cerulein (100 μg/kg) to establish HTG-AP and CER-AP, respectively. In HTG-AP, some mice received CQCQD (5.5 g/kg) gavage at 1, 5 or 9 h after disease induction. AP severity and related liver injury were determined by serological and histological parameters; and underlying mechanisms were identified by lipidomics and molecular biology. Molecular docking was used to identify key interactions between CQCQD compounds and metabolic enzymes, and subsequently validated in vitro in hepatocytes. RESULTS HTG-AP was associated with increased disease severity indices including augmented liver injury compared to CER-AP. CQCQD treatment reduced severity and liver injury of HTG-AP. Glycerophospholipid (GPL) metabolism was the most disturbed pathway in HTG-AP in comparison to HTG alone. In HTG-AP, the mRNA level of GPL enzymes involved in phosphocholine (PC) and phosphatidylethanolamine (PE) synthesis (Pcyt1a, Pcyt2, Pemt, and Lpcat) were markedly upregulated in the liver. Of the GPL metabolites, lysophosphatidylethanolamine LPE(16:0) in serum of HTG-AP was significantly elevated and positively correlated with the pancreas histopathology score (r = 0.65). In vitro, supernatant from Pcyt2-overexpressing hepatocytes co-incubated with LPE(16:0) or phospholipase A2 (a PC- and PE-hydrolyzing enzyme) alone induced pancreatic acinar cell death. CQCQD treatment downregulated PCYT1a and PCYT2 enzyme levels in the liver. Hesperidin and narirutin were identified top two CQCQD compounds with highest affinity docking to PCYT1a and PCYT2. Both hesperidin and narirutin reduced the level of some GPL metabolites in hepatocytes. CONCLUSION Liver-mediated GPL metabolism is excessively activated in HTG-AP with serum LPE(16:0) level correlating with disease severity. CQCQD reduces HTG-AP severity partially via modulating key enzymes in GPL metabolism pathway.
Collapse
Affiliation(s)
- Yongjian Wen
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuying Li
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tingting Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lijia Huang
- West China Biobank, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linbo Yao
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dan Deng
- West China Biobank, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenjuan Luo
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenhao Cai
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shaoqi Zhong
- West China Biobank, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Jin
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinmin Yang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiqi Wang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wen Wang
- Chinese Evidence-based Medicine Centre, and National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jing Xue
- Laboratory of Oncogenes and Related Genes, Stem Cell Research Centre, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Rajarshi Mukherjee
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool and Liverpool University Hospitals NHS Foundation Trust, Liverpoo,l L69 3GE, UK
| | - Jiwon Hong
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Anthony R Phillips
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - John A Windsor
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Robert Sutton
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool and Liverpool University Hospitals NHS Foundation Trust, Liverpoo,l L69 3GE, UK
| | - Fei Li
- Department of Pharmacy, Laboratory of Metabolomics and Drug-Induced Liver Injury, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Sun
- Chinese Evidence-based Medicine Centre, and National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Wei Huang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China; West China Biobank, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qing Xia
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
68
|
Zhang D, Shi C, Wang Y, Guo J, Gong Z. Metabolic Dysregulation and Metabolite Imbalances in Acute-on-chronic Liver Failure: Impact on Immune Status. J Clin Transl Hepatol 2024; 12:865-877. [PMID: 39440217 PMCID: PMC11491507 DOI: 10.14218/jcth.2024.00203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
Liver failure encompasses a range of severe clinical syndromes resulting from the deterioration of liver function, triggered by factors both within and outside the liver. While the definition of acute-on-chronic liver failure (ACLF) may vary by region, it is universally recognized for its association with multiorgan failure, a robust inflammatory response, and high short-term mortality rates. Recent advances in metabolomics have provided insights into energy metabolism and metabolite alterations specific to ACLF. Additionally, immunometabolism is increasingly acknowledged as a pivotal mechanism in regulating immune cell functions. Therefore, understanding the energy metabolism pathways involved in ACLF and investigating how metabolite imbalances affect immune cell functionality are crucial for developing effective treatment strategies for ACLF. This review methodically examined the immune and metabolic states of ACLF patients and elucidated how alterations in metabolites impact immune functions, offering novel perspectives for immune regulation and therapeutic management of liver failure.
Collapse
Affiliation(s)
- Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
69
|
Tong T, Zhu C, Farrell JJ, Khurshid Z, Martin ER, Pericak-Vance MA, Wang LS, Bush WS, Schellenberg GD, Haines JL, Qiu WQ, Lunetta KL, Farrer LA, Zhang X. Blood-derived mitochondrial DNA copy number is associated with Alzheimer disease, Alzheimer-related biomarkers and serum metabolites. Alzheimers Res Ther 2024; 16:234. [PMID: 39444005 PMCID: PMC11515778 DOI: 10.1186/s13195-024-01601-w] [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: 08/23/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Blood-derived mitochondrial DNA copy number (mtDNA-CN) is a proxy measurement of mitochondrial function in the peripheral and central systems. Abnormal mtDNA-CN not only indicates impaired mtDNA replication and transcription machinery but also dysregulated biological processes such as energy and lipid metabolism. However, the relationship between mtDNA-CN and Alzheimer disease (AD) is unclear. METHODS We performed two-sample Mendelian randomization (MR) using publicly available summary statistics from GWAS for mtDNA-CN and AD to investigate the causal relationship between mtDNA-CN and AD. We estimated mtDNA-CN using whole-genome sequence data from blood and brain samples of 13,799 individuals from the Alzheimer's Disease Sequencing Project. Linear and Cox proportional hazards models adjusting for age, sex, and study phase were used to assess the association of mtDNA-CN with AD. The association of AD biomarkers and serum metabolites with mtDNA-CN in blood was evaluated in Alzheimer's Disease Neuroimaging Initiative using linear regression. We conducted a causal mediation analysis to test the natural indirect effects of mtDNA-CN change on AD risk through the significantly associated biomarkers and metabolites. RESULTS MR analysis suggested a causal relationship between decreased blood-derived mtDNA-CN and increased risk of AD (OR = 0.68; P = 0.013). Survival analysis showed that decreased mtDNA-CN was significantly associated with higher risk of conversion from mild cognitive impairment to AD (HR = 0.80; P = 0.002). We also identified significant associations of mtDNA-CN with brain FDG-PET (β = 0.103; P = 0.022), amyloid-PET (β = 0.117; P = 0.034), CSF amyloid-β (Aβ) 42/40 (β=-0.124; P = 0.017), CSF t-Tau (β = 0.128; P = 0.015), p-Tau (β = 0.140; P = 0.008), and plasma NFL (β=-0.124; P = 0.004) in females. Several lipid species, amino acids, biogenic amines in serum were also significantly associated with mtDNA-CN. Causal mediation analyses showed that about a third of the effect of mtDNA-CN on AD risk was mediated by plasma NFL (P = 0.009), and this effect was more significant in females (P < 0.005). CONCLUSIONS Our study indicates that mtDNA-CN measured in blood is predictive of AD and is associated with AD biomarkers including plasma NFL particularly in females. Further, we illustrate that decreased mtDNA-CN possibly increases AD risk through dysregulation of mitochondrial lipid metabolism and inflammation.
Collapse
Affiliation(s)
- Tong Tong
- Bioinformatics Program, Boston University, Boston, MA, USA
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Congcong Zhu
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - John J Farrell
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Zainab Khurshid
- Bioinformatics Program, Boston University, Boston, MA, USA
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Eden R Martin
- Hussman Institute of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Margaret A Pericak-Vance
- Hussman Institute of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan L Haines
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Wei Qiao Qiu
- Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Lindsay A Farrer
- Bioinformatics Program, Boston University, Boston, MA, USA.
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
- Departments of Neurology and Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA.
- Boston University Chobanian & Avedisian School of Medicine, Biomedical Genetics E223, 72 East Concord Street, 02118, Boston, MA, USA.
| | - Xiaoling Zhang
- Bioinformatics Program, Boston University, Boston, MA, USA.
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
| |
Collapse
|
70
|
Chen M, Huang Z, Miao G, Ren J, Liu J, Roman MJ, Devereux RB, Fabsitz RR, Zhang Y, Umans JG, Cole SA, Kelly TN, Fiehn O, Zhao J. Longitudinal lipidomic profiles of left ventricular mass and left ventricular hypertrophy in American Indians. JCI Insight 2024; 9:e181172. [PMID: 39405119 PMCID: PMC11601894 DOI: 10.1172/jci.insight.181172] [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: 03/15/2024] [Accepted: 10/04/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUNDLeft ventricular hypertrophy (LVH) and dyslipidemia are strong, independent predictors for cardiovascular disease, but their relationship is less well studied. A longitudinal lipidomic profiling of left ventricular mass (LVM) and LVH is still lacking.METHODSUsing liquid chromatography-mass spectrometry (LC-MS), we repeatedly measured 1,542 lipids from 1,755 unique American Indians attending 2 exams (mean, 5 years apart). Cross-sectional associations of individual lipid species with LVM index (LVMI) were examined by generalized estimating equation (GEE), followed by replication in an independent biracial cohort (65% White, 35% Black). Baseline plasma lipids associated with LVH risk beyond traditional risk factors were identified by logistic GEE model in American Indians. Longitudinal associations between changes in lipids and changes in LVMI were examined by GEE, adjusting for baseline lipids, baseline LVMI, and covariates.RESULTSMultiple lipid species were significantly associated with LVMI or the risk of LVH in American Indians. Some lipids were confirmed in Black and White individuals. Moreover, some LVH-related lipids were inversely associated with risk of coronary heart disease (CHD). Longitudinal changes in several lipid species were significantly associated with changes in LVMI.CONCLUSIONAltered fasting plasma lipidome and its longitudinal change over time were significantly associated with LVMI and risk for LVH in American Indians. Our results offer insight into the role of individual lipid species in LV remodeling and risk of LVH, independent of known risk factors.FUNDINGThis study was supported by the NIH grant (R01DK107532). The Strong Heart Study has been funded in whole or in part with federal funds from the National Heart, Lung, and Blood Institute, NIH, Department of Health and Human Services, under contract nos. 75N92019D00027, 75N92019D00028, 75N92019D00029, and 75N92019D00030.
Collapse
Affiliation(s)
- Mingjing Chen
- Department of Epidemiology, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Guanhong Miao
- Department of Epidemiology, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jin Ren
- Department of Epidemiology, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jinling Liu
- Department of Epidemiology, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Mary J. Roman
- Division of Cardiology, Weill Cornell Medical College, New York, New York, USA
| | - Richard B. Devereux
- Division of Cardiology, Weill Cornell Medical College, New York, New York, USA
| | - Richard R. Fabsitz
- Missouri Breaks Industries Research Inc., Eagle Butte, South Dakota, USA
| | - Ying Zhang
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Jason G. Umans
- MedStar Health Research Institute, Hyattsville, Maryland, USA
- Georgetown-Howard Universities Center for Clinical and Translational Science, Washington DC, USA
| | - Shelley A. Cole
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Tanika N. Kelly
- Department of Medicine, University of Illinois Chicago, Chicago, Illinois, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, UCD, Davis, California, USA
| | - Jinying Zhao
- Department of Epidemiology, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
71
|
Yang J, Xiao H, Yao J, Zhang P, Yi B, Fang Z, Guo N, Guan Y, Zhang G. Integrated serum pharmacochemistry, 16S rDNA sequencing, and metabolomics to reveal the material basis and mechanism of Shouhui Tongbian capsule against diphenoxylate-induced slow transit constipation in rats. Chin Med 2024; 19:142. [PMID: 39394615 PMCID: PMC11468123 DOI: 10.1186/s13020-024-01015-8] [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: 11/08/2023] [Accepted: 09/26/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Slow transit constipation (STC) is highly prevalent and has rising incidence. Shouhui Tongbian capsule (SHTB) is a traditional Chinese Medicine formula with extensive and highly efficacious usage in STC treatment, however, its mechanism of action, especially the regulation of microbiome and lipid metabolites, remains unclear. METHODS After quality control of SHTB using LC‒MS to obtain its material basis, we tried to elucidate the cohesive modulatory network of SHTB against STC using hyphenated methods from microbiomics, lipidomics, mass spectrometry imaging (MSI) and molecular methods. RESULTS SHTB could repair intestinal barrier damage, reduce systemic inflammation and increase intestinal motility in a diphenoxylate-induced STC rat model. Based on 16S rDNA sequencing results, SHTB rehabilitated the abnormal changes in Alloprevotella, Coprococcus, Marvinbryantia, etc., which were associated with STC symptoms. Meanwhile, microbial functional prediction showed that lipid metabolism was improved with SHTB administration. The differential lipids, including fatty acids, lysophosphatidylcholine, phosphatidylcholine, sphingomyelin triglyceride and ceramide, that are closely related to STC disease and SHTB efficacy. Furthermore, SHTB significantly reversed the abnormal expression of these key target enzymes in colon samples, including CTP-phosphocholine cytidylyltransferase, CTP-phosphoethanolamine cytidylyltransferase, phosphatidic acid phosphatase, acid sphingomyelinase etc. CONCLUSIONS: Combined analysis demonstrated that SHTB reducing lipid accumulation and recovery of intestinal microbial homeostasis was the critical mechanism by which SHTB treats STC.
Collapse
Affiliation(s)
- Jiaying Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- College of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - He Xiao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Shandong, 273400, Linyi, China
| | - Jingchun Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Shandong, 273400, Linyi, China
| | - Pin Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Bojiao Yi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Zhengyu Fang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Na Guo
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Yongxia Guan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Shandong, 273400, Linyi, China.
| | - Guimin Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Shandong, 273400, Linyi, China.
| |
Collapse
|
72
|
Bushong A, Sepúlveda M, Scherer M, Valachovic AC, Neill CM, Horn S, Choi Y, Lee LS, Baloni P, Hoskins T. Effects of Perfluorinated Alkyl Substances (PFAS) on Amphibian Body and Liver Conditions: Is Lipid Metabolism Being Perturbed throughout Metamorphosis? TOXICS 2024; 12:732. [PMID: 39453152 PMCID: PMC11510839 DOI: 10.3390/toxics12100732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/29/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) may interact with peroxisome proliferator activated receptors (PPARs) and alter lipid homeostasis. Using Xenopus laevis, we investigated the effect of PFAS on (a) lipid homeostasis and whether this correlated to changes in body and hepatic condition; (b) the expression of hepatic genes regulated by PPAR; and (c) the hepatic lipidome. We chronically exposed tadpoles to 0.5 µg/L of either PFOS, PFHxS, PFOA, PFHxA, a binary mixture of PFOS and PFHxS (0.5 µg/L of each), or a control, from NF stage 52 through metamorphic climax. Growth, development, and survival were not affected, but we detected a sex-specific decrease in body condition at NF 66 (6.8%) and in hepatic condition (16.6%) across metamorphic climax for male tadpoles exposed to PFOS. We observed weak evidence for the transient downregulation of apolipoprotein-V (apoa5) at NF 62 in tadpoles exposed to PFHxA. Acyl-CoA oxidase 1 (acox1) was downregulated only in males exposed to PFHxS (Ln(Fold Change) = -0.54). We detected PFAS-specific downregulation of structural glycerophospholipids, while semi-quantitative profiling detected the upregulation in numerous glycerophospholipids, sphingomyelins, and diglycerides. Overall, our findings indicate that PFAS can induce sex-specific effects that change across larval development and metamorphosis. We demonstrate that PFAS alter lipid metabolism at environmentally relevant concentrations through divergent mechanisms that may not be related to PPARs, with an absence of effects on body condition, demonstrating the need for more molecular studies to elucidate mechanisms of PFAS-induced lipid dysregulation in amphibians and in other taxa.
Collapse
Affiliation(s)
- Anna Bushong
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN 47907, USA; (M.S.)
| | - Maria Sepúlveda
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN 47907, USA; (M.S.)
- Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
| | - Meredith Scherer
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN 47907, USA; (M.S.)
| | - Abigail C. Valachovic
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN 47907, USA; (M.S.)
| | - C. Melman Neill
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN 47907, USA; (M.S.)
| | - Sophia Horn
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN 47907, USA; (M.S.)
| | - Youn Choi
- Department of Agronomy and Environmental & Ecological Engineering, Interdisciplinary Ecological Sciences and Engineering, Purdue University, West Lafayette, IN 47907, USA; (Y.C.); (L.S.L.)
| | - Linda S. Lee
- Department of Agronomy and Environmental & Ecological Engineering, Interdisciplinary Ecological Sciences and Engineering, Purdue University, West Lafayette, IN 47907, USA; (Y.C.); (L.S.L.)
| | - Priyanka Baloni
- College of Health Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Tyler Hoskins
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN 47907, USA; (M.S.)
| |
Collapse
|
73
|
Turpin J, Wadolowski S, Tambo W, Kim D, Al Abed Y, Sciubba DM, Becker LB, Ledoux D, Kim J, Powell K, Li C. Exploring Lysophosphatidylcholine as a Biomarker in Ischemic Stroke: The Plasma-Brain Disjunction. Int J Mol Sci 2024; 25:10649. [PMID: 39408978 PMCID: PMC11477326 DOI: 10.3390/ijms251910649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Lipids and their bioactive metabolites, notably lysophosphatidylcholine (LPC), are increasingly important in ischemic stroke research. Reduced plasma LPC levels have been linked to stroke occurrence and poor outcomes, positioning LPC as a potential prognostic or diagnostic marker. Nonetheless, the connection between plasma LPC levels and stroke severity remains unclear. This study aimed to elucidate this relationship by examining plasma LPC levels in conjunction with brain LPC levels to provide a deeper understanding of the underlying mechanisms. Adult male Sprague-Dawley rats underwent transient middle cerebral artery occlusion and were randomly assigned to different groups (sham-operated, vehicle, LPC supplementation, or LPC inhibition). We measured multiple LPC species in the plasma and brain, alongside assessing sensorimotor dysfunction, cerebral perfusion, lesion volume, and markers of BBB damage, inflammation, apoptosis, and oxidative stress. Among five LPC species, plasma LPC(16:0) and LPC(18:1) showed strong correlations with sensorimotor dysfunction, lesion severity, and mechanistic biomarkers in the rat stroke model. Despite notable discrepancies between plasma and brain LPC levels, both were strongly linked to functional outcomes and mechanistic biomarkers, suggesting that LPC's prognostic value is retained extracranially. This study advances the understanding of LPC as a blood marker in ischemic stroke and highlights directions for future research to further elucidate its association with stroke severity, particularly through investigations in more clinically representative models.
Collapse
Affiliation(s)
- Justin Turpin
- Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
| | - Steven Wadolowski
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
| | - Willians Tambo
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, New York, NY 11030, USA
| | - Daniel Kim
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA
| | - Yousef Al Abed
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, New York, NY 11030, USA
| | - Daniel M. Sciubba
- Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Lance B. Becker
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, New York, NY 11030, USA
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
| | - David Ledoux
- Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Junhwan Kim
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
| | - Keren Powell
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
| | - Chunyan Li
- Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, NY 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, New York, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
74
|
Bi Y, Ying X, Chen W, Wu J, Kong C, Hu W, Fang S, Yu J, Zhai M, Jiang C, Chen M, Shen L, Ji J, Tu J. Glycerophospholipid-driven lipid metabolic reprogramming as a common key mechanism in the progression of human primary hepatocellular carcinoma and cholangiocarcinoma. Lipids Health Dis 2024; 23:326. [PMID: 39354487 PMCID: PMC11443871 DOI: 10.1186/s12944-024-02298-4] [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: 07/18/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
Metabolic reprogramming, a key mechanism regulating the growth and recurrence of hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), still lacks effective clinical strategies for its integration into the precise screening of primary liver cancer. This study utilized ultra-high-performance liquid chromatography with quadrupole time-of-flight mass spectrometry to conduct a comprehensive, non-targeted metabolomics analysis, revealing significant upregulation of lipid metabolites such as phosphatidylcholine and lysophosphatidylcholine in patients with HCC and CCA, particularly within the glycerophospholipid metabolic pathway. Hematoxylin and eosin and immunohistochemical staining demonstrated marked upregulation of phospholipase A2 in tumor tissues, further emphasizing the potential of lipid metabolism as a therapeutic target and its important part in the course of cancer. This work provides a new viewpoint for addressing the clinical challenges associated with HCC and CCA, laying the groundwork for the broad application of early diagnosis and personalized treatment strategies, and ultimately aiming to provide tailored and precise therapeutic options for patients.
Collapse
Affiliation(s)
- Yanran Bi
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Xihui Ying
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Wanbin Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Jiahao Wu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Chunli Kong
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Weiming Hu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Shiji Fang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Junchao Yu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Mengqian Zhai
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Chengli Jiang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Lin Shen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China.
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui, 323000, China.
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China.
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China.
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China.
| | - Jianfei Tu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China.
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China.
| |
Collapse
|
75
|
Yin C, Liu L, Xu D, Li M, Li M, Qin Y, Zhang B, Sun Y, Liu Y, Xiao Y. Integrative metagenomic and lipidomic analyses reveal alterations in children with obesity and after lifestyle intervention. Front Nutr 2024; 11:1423724. [PMID: 39318384 PMCID: PMC11420138 DOI: 10.3389/fnut.2024.1423724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Background Despite emerging evidence linking alterations in gut microbiota to childhood obesity, the metabolic mechanisms linking gut microbiota to the lipid profile during childhood obesity and weight loss remain poorly understood. Methodology In this study, children with obesity were treated with lifestyle weight loss therapy. Metagenomics association studies and serum untargeted lipidomics analyses were performed in children with obesity and healthy controls before and after weight loss. Main findings We identified alterations in gut microbiota associated with childhood obesity, as well as variations in circulating metabolite concentrations. Children with obesity showed significant decreases in the levels of s-Rothia_kristinae and s-Enterobacter_roggenkampii, alongsige elevated levels of s-Clostridiales_bacterium_Marseille-P5551. Following weight loss, the levels of s-Streptococcus_infantarius and s-Leuconostoc_citreum increased by factors of 3.354 and 1.505, respectively, in comparison to their pre-weight loss levels. Correlation analyses indicated a significant positive relationship between ChE(2:0) levels and both with s-Lachnospiraceae_bacterium_TF09-5 and fasting glucose levels. CoQ8 levels were significantly negatively correlated with s-Rothia_kristinae and HOMA-IR. Conclusion We linked altered gut microbiota and serum lipid levels in children with obesity to clinical indicators, indicating a potential impact on glucose metabolism via lipids. This study contributes to understanding the mechanistic relationship between altered gut microbiota and childhood obesity and weight loss, suggesting gut microbiome as a promising target for intervention. Clinical trial registration https://www.chictr.org.cn/showproj.html?proj=178971, ChiCTR2300072179.
Collapse
Affiliation(s)
- Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lujie Liu
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Dong Xu
- Department of Pediatrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Li
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Min Li
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yujie Qin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bei Zhang
- Department of Pediatrics, Luoyang Central Hospital, Luoyang, China
| | - Yongfa Sun
- Department of Pediatrics, Luoyang Central Hospital, Luoyang, China
| | - Yuesheng Liu
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yanfeng Xiao
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
76
|
Noone DP, Isendoorn MME, Hamers SMWR, Keizer ME, Wulffelé J, van der Velden TT, Dijkstra DJ, Trouw LA, Filippov DV, Sharp TH. Structural basis for surface activation of the classical complement cascade by the short pentraxin C-reactive protein. Proc Natl Acad Sci U S A 2024; 121:e2404542121. [PMID: 39240968 PMCID: PMC11406272 DOI: 10.1073/pnas.2404542121] [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: 03/15/2024] [Accepted: 08/04/2024] [Indexed: 09/08/2024] Open
Abstract
Human C-reactive protein (CRP) is a pentameric complex involved in immune defense and regulation of autoimmunity. CRP is also a therapeutic target, with both administration and depletion of serum CRP being pursued as a possible treatment for autoimmune and cardiovascular diseases, among others. CRP binds to phosphocholine (PC) moieties on membranes to activate the complement system via the C1 complex, but it is unknown how CRP, or any pentraxin, binds to C1. Here, we present a cryoelectron tomography (cryoET)-derived structure of CRP bound to PC ligands and the C1 complex. To gain control of CRP binding, a synthetic mimotope of PC was synthesized and used to decorate cell-mimetic liposome surfaces. Structure-guided mutagenesis of CRP yielded a fully active complex able to bind PC-coated liposomes that was ideal for cryoET and subtomogram averaging. In contrast to antibodies, which form Fc-mediated hexameric platforms to bind and activate the C1 complex, CRP formed rectangular platforms assembled from four laterally associated CRP pentamers that bind only four of the six available globular C1 head groups. Potential residues mediating lateral association of CRP were identified from interactions between unit cells in existing crystal structures, which rationalized previously unexplained mutagenesis data regarding CRP-mediated complement activation. The structure also enabled interpretation of existing biochemical data regarding interactions mediating C1 binding and identified additional residues for further mutagenesis studies. These structural data therefore provide a possible mechanism for regulation of complement by CRP, which limits complement progression and has consequences for how the innate immune system influences autoimmunity.
Collapse
Affiliation(s)
- Dylan P. Noone
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
| | - Marjolein M. E. Isendoorn
- Leiden Institute of Chemistry, Gorlaeus Laboratory, Leiden University, 2333 CCLeiden, The Netherlands
| | - Sebastiaan M. W. R. Hamers
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
| | - Mariska E. Keizer
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
| | - Jip Wulffelé
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
| | - Tijn T. van der Velden
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
| | - Douwe J. Dijkstra
- Department of Immunology, Leiden University Medical Center, 2333 ZALeiden, The Netherlands
| | - Leendert A. Trouw
- Department of Immunology, Leiden University Medical Center, 2333 ZALeiden, The Netherlands
| | - Dmitri V. Filippov
- Leiden Institute of Chemistry, Gorlaeus Laboratory, Leiden University, 2333 CCLeiden, The Netherlands
| | - Thomas H. Sharp
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
- School of Biochemistry, University of Bristol, BristolBS8 1TD, United Kingdom
| |
Collapse
|
77
|
Banton S, Singh P, Seymour DJ, Saunders-Blades J, Shoveller AK. Postprandial Plasma and Whole Blood Amino Acids Are Largely Indicative of Dietary Amino Acids in Adult Dogs Consuming Diets with Increasing Whole Pulse Ingredient Inclusion. J Nutr 2024; 154:2655-2669. [PMID: 39025332 PMCID: PMC11393166 DOI: 10.1016/j.tjnut.2024.07.023] [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: 02/27/2024] [Revised: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Pulse ingredients often replace grains in grain-free dog diets owing to their high-protein content. However, research to ascertain the benefit of this modification is limited. OBJECTIVES This study aimed to correlate food compounds in 1 corn-inclusive control diet and 3 grain-free diets with increasing inclusions of whole pulses (≤45%; Pulse15, Pulse30, and Pulse45), formulated to meet similar macronutrient and micronutrient targets with postprandial amino acids (AAs) in healthy dogs >20 wk. METHODS Diets were analyzed for biochemical compounds using tandem mass spectrometry. Twenty-eight outdoor-housed, healthy, adult Siberian Huskies were allocated to diet, and meal responses were analyzed at baseline and weeks 2, 4, 8, 16, and 20 with samples collected at fasted and 15, 30, 60, 90, 120, and 180 min after meal presentation. Blood AAs were analyzed by ultra performance liquid chromatography and differences across week, treatment, and time postmeal were analyzed in SAS Studio. Partial least squares regression was performed in SAS Studio using biochemical compounds in the diet as predictor variables and blood AAs as response variables. RESULTS In plasma, Pulse45 had ∼32% greater postprandial Asn than Pulse15, and the control diet had ∼34% greater postprandial Leu and ∼35% greater Pro than Pulse15 (P < 0.05). In whole blood, Pulse30 had ∼23% greater postprandial Lys than the control diet, and the control diet had ∼21% greater postprandial Met and ∼18% greater Pro than Pulse45 and Pulse30, respectively (P < 0.05). Several phospholipids were correlated with postprandial AAs. Compounds in the urea cycle and glycine and serine metabolism were more enriched (P < 0.05) in plasma and whole blood, respectively. CONCLUSIONS In macronutrient-balanced and micronutrient-balanced canine diets that differ in their inclusion of corn-derived compared with pulse-derived ingredients, postprandial changes in circulating AAs are largely indicative of the dietary AAs. This helps further our understanding of AA metabolism in healthy dogs fed grain-free diets.
Collapse
Affiliation(s)
- Sydney Banton
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario, Canada
| | - Pawanpreet Singh
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario, Canada
| | - Dave J Seymour
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario, Canada; Trouw Nutrition R&D, Amersfoort, Netherlands
| | | | - Anna K Shoveller
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
78
|
Lee DY. Emerging Circulating Biomarkers for Enhanced Cardiovascular Risk Prediction. J Lipid Atheroscler 2024; 13:262-279. [PMID: 39355403 PMCID: PMC11439747 DOI: 10.12997/jla.2024.13.3.262] [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: 01/04/2024] [Revised: 04/12/2024] [Accepted: 06/06/2024] [Indexed: 10/03/2024] Open
Abstract
Cardiovascular disease (CVD) continues to be the primary cause of mortality worldwide, underscoring the importance of identifying additional cardiovascular risk factors. The consensus is that lipid levels alone do not fully reflect the status of atherosclerosis, thus necessitating extensive research on cardiovascular biomarkers. This review encompasses a wide spectrum of methodologies for identifying novel risk factors or biomarkers for CVD. Inflammation, oxidative stress, plaque instability, cardiac remodeling, and fibrosis play pivotal roles in CVD pathogenesis. We introduce and discuss several promising biomarkers-namely, osteocalcin, angiogenin, lipoprotein-associated phospholipase A2, growth differentiation factor 15, galectin-3, growth stimulation expressed gene 2, and microRNAs, all of which have potential implications in the assessment and management of cardiovascular risk.
Collapse
Affiliation(s)
- Da Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
79
|
Lin WL, Chien MM, Patchara S, Wang W, Faradina A, Huang SY, Tung TH, Tsai CS, Skalny AV, Tinkov AA, Chang CC, Chang JS. Essential trace element and phosphatidylcholine remodeling: Implications for body composition and insulin resistance. J Trace Elem Med Biol 2024; 85:127479. [PMID: 38878466 DOI: 10.1016/j.jtemb.2024.127479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Recent studies indicated that bioactive lipids of phosphatidylcholines (PCs) and lysophosphatidylcholines (LysoPCs) predict unhealthy metabolic phenotypes, but results remain inconsistent. To fill this knowledge gap, we investigated whether essential trace elements affect PC-Lyso PC remodeling pathways and the risk of insulin resistance (IR). METHODS Anthropometric and blood biochemical data (glucose, insulin, and lipoprotein-associated phospholipase A2 (Lp-PLA2)) were obtained from 99 adults. Blood essential/probably essential trace elements and lipid metabolites were respectively measured by inductively coupled plasma mass spectrometry (ICP-MS), and ultra-performance liquid chromatography-mass spectrometry (UPLC-MS). RESULT AND CONCLUSION Except for LysoPC (O-18:0/0:0), an inverse V shape was observed between body weight and PC and LysoPC species. A Pearson correlation analysis showed that essential/probably-essential metals (Se, Cu, and Ni: r=-0.4∼-0.7) were negatively correlated with PC metabolites but positively correlated with LysoPC (O-18:0/0:0) (Se, Cu, and Ni: r=0.85-0.64). Quantile-g computation showed that one quantile increase in essential metals was associated with a 2.16-fold increase in serum Lp-PLA2 (β=2.16 (95 % confidence interval (CI): 0.34, 3.98), p=0.023), which are key enzymes involved in PC/Lyso PC metabolism. An interactive analysis showed that compared to those with the lowest levels (reference), individuals with the highest levels of serum PCs (pooled, M2) and the lowest essential/probably essential metals (M1) were associated with a healthier body composition and had a 76 % decreased risk of IR (odds ratio (OR)=0.24 (95 % CI: 0.06, 0.90), p<0.05). In contrast, increased exposure to LysoPC(O-18:0/0:0) (M2) and essential metals (M2) exhibited an 8.22-times highest risk of IR (OR= 8.22 (2.07, 32.57), p<0.05) as well as an altered body composition. In conclusion, overexposure to essential/probably essential trace elements may promote an unhealthy body weight and IR through modulating PC/LysoPC remodeling pathways.
Collapse
Affiliation(s)
- Wen-Ling Lin
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan, ROC
| | - Mu-Ming Chien
- Department of Pediatrics, Taipei Medical University Hospital, Taipei 11031, Taiwan, ROC
| | - Sangopas Patchara
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan, ROC
| | - Weu Wang
- Division of Digestive Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 11301, Taiwan, ROC; Department of Surgery, College of Medicine, Taipei Medical University, Taipei 11301, Taiwan, ROC
| | - Amelia Faradina
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan, ROC
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan, ROC; Center for Reproductive Medicine & Sciences, Taipei Medical University Hospital, Taipei 11031, Taiwan, ROC; Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan, ROC
| | - Te-Hsuan Tung
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Tri-service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan, ROC; Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 114202, Taiwan, ROC
| | - Anatoly V Skalny
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - Alexey A Tinkov
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, Russia
| | - Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taiwan, ROC; Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Collage of Medicine, Taipei Medical University, Taipei 11031, Taiwan, ROC
| | - Jung-Su Chang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan, ROC; Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan, ROC; Chinese Taipei Society for the Study of Obesity (CTSSO), Taipei 11031, Taiwan, ROC; TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 11031, Taiwan, ROC.
| |
Collapse
|
80
|
Yadav P, Beura SK, Panigrahi AR, Kulkarni PP, Yadav MK, Munshi A, Singh SK. Lysophosphatidylcholine induces oxidative stress and calcium-mediated cell death in human blood platelets. Cell Biol Int 2024; 48:1266-1284. [PMID: 38837523 DOI: 10.1002/cbin.12192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 06/07/2024]
Abstract
Platelets are essential component of circulation that plays a major role in hemostasis and thrombosis. During activation and its demise, platelets release platelet-derived microvesicles, with lysophosphatidylcholine (LPC) being a prominent component in their lipid composition. LPC, an oxidized low-density lipoprotein, is involved in cellular metabolism, but its higher level is implicated in pathologies like atherosclerosis, diabetes, and inflammatory disorders. Despite this, its impact on platelet function remains relatively unexplored. To address this, we studied LPC's effects on washed human platelets. A multimode plate reader was employed to measure reactive oxygen species and intracellular calcium using H2DCF-DA and Fluo-4-AM, respectively. Flow cytometry was utilized to measure phosphatidylserine expression, mitochondrial membrane potential (ΔΨm), and mitochondrial permeability transition pore (mPTP) formation using FITC-Annexin V, JC-1, and CoCl2/calcein-AM, respectively. Additionally, platelet morphology and its ultrastructure were observed via phase contrast and electron microscopy. Sonoclot and light transmission aggregometry were employed to examine fibrin formation and platelet aggregation, respectively. The findings demonstrate that LPC induced oxidative stress and increased intracellular calcium in platelets, resulting in increased phosphatidylserine expression and reduced ΔΨm. LPC triggered caspase-independent platelet death and mPTP opening via cytosolic and mitochondrial calcium, along with microvesiculation and reduced platelet counts. LPC increased the platelet's size, adopting a balloon-shaped morphology, causing membrane fragmentation and releasing its cellular contents, while inducing a pro-coagulant phenotype with increased fibrin formation and reduced integrin αIIbβ3 activation. Conclusively, this study reveals LPC-induced oxidative stress and calcium-mediated platelet death, necrotic in nature with pro-coagulant properties, potentially impacting inflammation and repair mechanisms during vascular injury.
Collapse
Affiliation(s)
- Pooja Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Samir K Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Abhishek R Panigrahi
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Paresh P Kulkarni
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mithlesh K Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Ghudda, Bathinda, India
| | - Sunil K Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| |
Collapse
|
81
|
Huang J, Liu M, Zhang H, Sun G, Furey A, Rahman P, Zhai G. Multi-Omics Integrative Analyses Identified Two Endotypes of Hip Osteoarthritis. Metabolites 2024; 14:480. [PMID: 39330487 PMCID: PMC11434176 DOI: 10.3390/metabo14090480] [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: 07/13/2024] [Revised: 08/10/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
(1) Background: Osteoarthritis (OA) is a heterogeneous disorder, and subgroup classification of OA remains elusive. The aim of our study was to identify endotypes of hip OA and investigate the altered pathways in the different endotypes. (2) Methods: Metabolomic profiling and genome-wide genotyping were performed on fasting blood. Transcriptomic profiling was performed on RNA extracted from cartilage samples. Machine learning methods were used to identify endotypes of hip OA. Pathway analysis was used to identify the altered pathways between hip endotypes and controls. GWAS was performed on each of the identified metabolites. Transcriptomic data was used to examine the expression levels of identified genes in cartilage. (3) Results: 180 hip OA patients and 120 OA-free controls were classified into three clusters based on metabolomic data. The combination of arginine, ornithine, and the average value of 7 lysophosphatidylcholines had an area under the curve (AUC) of 0.97 (95% CI: 0.96-0.99) to discriminate hip OA from controls, and the combination of γ-aminobutyric acid, spermine, aconitic acid, and succinic acid had an AUC of 0.96 (95% CI: 0.94-0.99) to distinguish two hip OA endotypes. GWAS identified 236 SNPs to be associated with identified metabolites at GWAS significance level. Pro-inflammatory cytokine levels were significantly different between two endotypes (all p < 0.05). (4) Conclusions: Hip OA could be classified into two distinct molecular endotypes. The primary differences between the two endotypes involve changes in pro-inflammatory factors and energy metabolism.
Collapse
Affiliation(s)
- Jingyi Huang
- Human Genetics & Genomics, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Ming Liu
- Human Genetics & Genomics, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Hongwei Zhang
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Guang Sun
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Andrew Furey
- Discipline of Surgery, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
- Office of the Premier, Government of Newfoundland & Labrador, St. John's, NL A1B 4J6, Canada
| | - Proton Rahman
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Guangju Zhai
- Human Genetics & Genomics, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| |
Collapse
|
82
|
Liu S, Wang Y, Ying L, Li H, Zhang K, Liang N, Luo G, Xiao L. Quercetin Mitigates Lysophosphatidylcholine (LPC)-Induced Neutrophil Extracellular Traps (NETs) Formation through Inhibiting the P2X7R/P38MAPK/NOX2 Pathway. Int J Mol Sci 2024; 25:9411. [PMID: 39273358 PMCID: PMC11395007 DOI: 10.3390/ijms25179411] [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: 07/18/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are three-dimensional reticular structures that release chromatin and cellular contents extracellularly upon neutrophil activation. As a novel effector mechanism of neutrophils, NETs possess the capacity to amplify localized inflammation and have been demonstrated to contribute to the exacerbation of various inflammatory diseases, including cardiovascular diseases and tumors. It is suggested that lysophosphatidylcholine (LPC), as the primary active component of oxidized low-density lipoprotein, represents a significant risk factor for various inflammatory diseases, such as cardiovascular diseases and neurodegenerative diseases. However, the specific mechanism of NETs formation induced by LPC remains unclear. Quercetin has garnered considerable attention due to its anti-inflammatory properties, serving as a prevalent flavonoid in daily diet. However, little is currently known about the underlying mechanisms by which quercetin inhibits NETs formation and alleviates associated diseases. In our study, we utilized LPC-treated primary rat neutrophils to establish an in vitro model of NETs formation, which was subsequently subjected to treatment with a combination of quercetin or relevant inhibitors/activators. Compared to the control group, the markers of NETs and the expression of P2X7R/P38MAPK/NOX2 pathway-associated proteins were significantly increased in cells treated with LPC alone. Quercetin intervention decreased the LPC-induced upregulation of the P2X7R/P38MAPK/NOX2 pathway and effectively reduced the expression of NETs markers. The results obtained using a P2X7R antagonist/activator and P38MAPK inhibitor/activator support these findings. In summary, quercetin reversed the upregulation of the LPC-induced P2X7R/P38MAPK/NOX2 pathway, further mitigating NETs formation. Our study investigated the potential mechanism of LPC-induced NETs formation, elucidated the inhibitory effect of quercetin on NETs formation, and offered new insights into the anti-inflammatory properties of quercetin.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gang Luo
- Xiangya School of Public Health, Central South University, Changsha 410013, China; (S.L.); (Y.W.); (L.Y.); (H.L.); (K.Z.); (N.L.)
| | - Lin Xiao
- Xiangya School of Public Health, Central South University, Changsha 410013, China; (S.L.); (Y.W.); (L.Y.); (H.L.); (K.Z.); (N.L.)
| |
Collapse
|
83
|
Ahern MM, Artegoitia VM, Bosviel R, Newman JW, Keim NL, Krishnan S. Fat burning capacity in a mixed macronutrient meal protocol does not reflect metabolic flexibility in women who are overweight or obese. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.29.24312791. [PMID: 39252930 PMCID: PMC11383504 DOI: 10.1101/2024.08.29.24312791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Introduction Metabolic flexibility, the ability to switch from glucose to fat as a fuel source, is considered a marker of metabolic health. Higher fat oxidation is often associated with greater flexibility and insulin sensitivity, while lower fat oxidation is linked to metabolic inflexibility and insulin resistance. However, our study challenges the universal validity of this relationship, uncovering a more nuanced understanding of the complex interplay between fuel source switching and fat oxidation, especially in the presence of insulin resistance. Methods In an 8-week controlled feeding intervention, overweight to obese women with insulin resistance (as defined by McAuley's index) were randomized to consume either a diet based on the Dietary Guidelines for Americans 2010 (DGA) or a 'Typical' American Diet (TAD), n = 22 each. Participants were given a high-fat mixed macronutrient challenge test (MMCT) (60% fat, 28% carbohydrates, and 12% protein) at weeks 0, 2, and 8. Plasma lipids, metabolome, and lipidome were measured at 0, 0.5, 3, and 6h postprandial (PP); substrate oxidation measures were also recorded at 0,1 3, and 6h PP. Metabolic flexibility was evaluated as the change in fat oxidation from fasting to PP. Mixed model and multivariate analyses were used to evaluate the effect of diet on these outcomes, and to identify variables of interest to metabolic flexibility. Results Intervention diets (DGA and TAD) did not differentially affect substrate oxidation or metabolic flexibility, and equivalence tests indicated that groups could be combined for subsequent analyses. Participants were classified into three groups based on the % of consumed MMCT fat was oxidized in the 6h post meal period at weeks 0, 2 and 8. Low fat burners (LB, n = 6, burned <30% of fat in MMCT) and high fat burners (HB, n = 7, burned > 40% of fat in MMCT) at all weeks. Compared to LB, HB group had higher fat mass, total mass, lean mass, BMI, lower HDLc and lower RER (p < 0.05), but not different % body fat or % lean mass. During week 0, at 1h PP, LB had an increase in % fat oxidation change from 0h compared to HB (p<0.05), suggesting higher metabolic flexibility. This difference disappeared later in the PP phase, and we did not detect this beyond week 0. Partial least squares discriminant analysis (PLSDA (regular and repeated measures (sPLSDA)) models identified that LB group, in the late PP phase, was associated with higher rates of disappearance of acylcarnitines (AC) and lysophosphatidylcholines (LPC) from plasma (Q2: 0.20, R2X: 0.177, R2Y: 0.716). Conclusion In women with insulin resistance, a high fat burning capacity does not imply high metabolic flexibility, and not all women with insulin resistance are metabolically inflexible. LPCs and ACs are promising biomarkers of metabolic flexibility.
Collapse
Affiliation(s)
- Mary M. Ahern
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson AZ 85721
| | - Virginia M. Artegoitia
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA 95616, USA
| | - Rémy Bosviel
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
| | - John W. Newman
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA 95616, USA
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
- Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
| | - Nancy L. Keim
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA 95616, USA
- Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
| | - Sridevi Krishnan
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson AZ 85721
| |
Collapse
|
84
|
Xiong Y, Li X, Liu J, Luo P, Zhang H, Zhou H, Ling X, Zhang M, Liang Y, Chen Q, Xing C, Li F, Miao J, Shen W, Zhou S, Wang X, Hou FF, Liu Y, Ma K, Zhao AZ, Zhou L. Omega-3 PUFAs slow organ aging through promoting energy metabolism. Pharmacol Res 2024; 208:107384. [PMID: 39209083 DOI: 10.1016/j.phrs.2024.107384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Energy metabolism disorder, mainly exhibiting the inhibition of fatty acid degradation and lipid accumulation, is highly related with aging acceleration. However, the intervention measures are deficient. Here, we reported Omega-3 polyunsaturated fatty acids (Omega-3 PUFAs), especially EPA, exerted beneficial effects on maintaining energy metabolism and lipid homeostasis to slow organ aging. As the endogenous agonist of peroxisome proliferator-activated receptor α (PPARα), Omega-3 PUFAs significantly boosted fatty acid β-oxidation and ATP production in multiple aged organs. Consequently, Omega-3 PUFAs effectively inhibited age-related pathological changes, preserved organ function, and retarded aging process. The beneficial effects of Omega-3 PUFAs were also testified in mfat-1 transgenic mice, which spontaneously generate abundant endogenous Omega-3 PUFAs. In conclusion, our study innovatively demonstrated Omega-3 PUFAs administration in diet slow aging through promoting energy metabolism. The supplement of Omega-3 PUFAs or fat-1 transgene provides a promising therapeutic approach to promote healthy aging in the elderly.
Collapse
Affiliation(s)
- Yabing Xiong
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Nephrology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaolong Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiafeng Liu
- Department of Nephrology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Pei Luo
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haixia Zhang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hong Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xian Ling
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meijia Zhang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Liang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiurong Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chaofeng Xing
- Department of Endocrinology, Shunde Hospital of Southern Medical University, No.1 Jiazi Road, Foshan, Guangdong Province, China
| | - Fanghong Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Jinhua Miao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiwei Shen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxu Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kunling Ma
- Department of Nephrology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Allan Zijian Zhao
- Department of Endocrinology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
85
|
Li Y, Wang Z, Kong M, Yong Y, Yang X, Liu C. The role of GZMA as a target of cysteine and biomarker in Alzheimer's disease, pelvic organ prolapse, and tumor progression. Front Pharmacol 2024; 15:1447605. [PMID: 39228516 PMCID: PMC11368878 DOI: 10.3389/fphar.2024.1447605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Objective: This study aims to investigate how changes in peripheral blood metabolites in Alzheimer's Disease (AD) patients affect the development of Pelvic Organ Prolapse (POP) using a multi-omics approach. We specifically explore the interactions of signaling pathways, gene expression, and protein-metabolite interactions, with a focus on GZMA and cysteine in age-related diseases. Methods: This study utilized multi-omics analysis, including metabolomics and transcriptomics, to evaluate the perturbations in peripheral blood metabolites and their effect on POP in AD patients. Additionally, a comprehensive pan-cancer and immune infiltration analysis was performed on the core targets of AD combined with POP, exploring their potential roles in tumor progression and elucidating their pharmacological relevance to solid tumors. Results: We identified 47 differential metabolites linked to 9 significant signaling pathways, such as unsaturated fatty acid biosynthesis and amino acid metabolism. A thorough gene expression analysis revealed numerous differentially expressed genes (DEGs), with Gene Set Enrichment Analysis (GSEA) showing significant changes in gene profiles of AD and POP. Network topology analysis highlighted central nodes in the AD-POP co-expressed genes network. Functional analyses indicated involvement in critical biological processes and pathways. Molecular docking studies showed strong interactions between cysteine and proteins PTGS2 and GZMA, and molecular dynamics simulations confirmed the stability of these complexes. In vitro validation demonstrated that cysteine reduced ROS levels and protected cell viability. GZMA was widely expressed in various cancers, associated with immune cells, and correlated with patient survival prognosis. Conclusion: Multi-omics analysis revealed the role of peripheral blood metabolites in the molecular dynamics of AD and their interactions with POP. This study identified potential biomarkers and therapeutic targets, emphasizing the effectiveness of integrative approaches in treating AD and POP concurrently. The findings highlight the need for in-depth research on novel targets and biomarkers to advance therapeutic strategies.
Collapse
Affiliation(s)
- Yan Li
- Department of Gynecology and Obstetrics, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
- Department of Gynecology and Obstetrics, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhuo Wang
- Department of Gynecology and Obstetrics, Ningxia Medical University, Yinchuan, China
| | - Min Kong
- Department of Gynecology and Obstetrics, Ningxia Medical University, Yinchuan, China
| | - Yuanyuan Yong
- Department of Gynecology and Obstetrics, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xin Yang
- Department of Gynecology and Obstetrics, Ningxia Medical University, Yinchuan, China
| | - Chongdong Liu
- Department of Gynecology and Obstetrics, Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
86
|
Hung SC, Chan TF, Chan HC, Wu CY, Chan ML, Jhuang JY, Tan JQ, Mei JB, Law SH, Ponnusamy VK, Chan HC, Ke LY. Lysophosphatidylcholine Impairs the Mitochondria Homeostasis Leading to Trophoblast Dysfunction in Gestational Diabetes Mellitus. Antioxidants (Basel) 2024; 13:1007. [PMID: 39199251 PMCID: PMC11351454 DOI: 10.3390/antiox13081007] [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: 07/05/2024] [Revised: 08/10/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is a common pregnancy disorder associated with an increased risk of pre-eclampsia and macrosomia. Recent research has shown that the buildup of excess lipids within the placental trophoblast impairs mitochondrial function. However, the exact lipids that impact the placental trophoblast and the underlying mechanism remain unclear. GDM cases and healthy controls were recruited at Kaohsiung Medical University Hospital. The placenta and cord blood were taken during birth. Confocal and electron microscopy were utilized to examine the morphology of the placenta and mitochondria. We determined the lipid composition using liquid chromatography-mass spectrometry in data-independent analysis mode (LC/MSE). In vitro studies were carried out on choriocarcinoma cells (JEG3) to investigate the mechanism of trophoblast mitochondrial dysfunction. Results showed that the GDM placenta was distinguished by increased syncytial knots, chorangiosis, lectin-like oxidized low-density lipoprotein (LDL) receptor-1 (LOX-1) overexpression, and mitochondrial dysfunction. Lysophosphatidylcholine (LPC) 16:0 was significantly elevated in the cord blood LDL of GDM patients. In vitro, we demonstrated that LPC dose-dependently disrupts mitochondrial function by increasing reactive oxygen species (ROS) levels and HIF-1α signaling. In conclusion, highly elevated LPC in cord blood plays a pivotal role in GDM, contributing to trophoblast impairment and pregnancy complications.
Collapse
Affiliation(s)
- Shao-Chi Hung
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
| | - Te-Fu Chan
- Graduate Institute of Medicine, College of Medicine & Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
| | - Hsiu-Chuan Chan
- PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (H.-C.C.); (V.K.P.)
| | - Chia-Ying Wu
- The Master Program of AI Application in Health Industry, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
| | - Mei-Lin Chan
- Division of Thoracic Surgery, Department of Surgery, MacKay Memorial Hospital, MacKay Medical College, Taipei 104217, Taiwan;
- Department of Medicine, MacKay Medical College, New Taipei 252005, Taiwan;
| | - Jie-Yang Jhuang
- Department of Medicine, MacKay Medical College, New Taipei 252005, Taiwan;
- Department of Pathology, Mackay Memorial Hospital, Tamsui Branch, New Taipei 251404, Taiwan
| | - Ji-Qin Tan
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
| | - Jia-Bin Mei
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
| | - Shi-Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
| | - Vinoth Kumar Ponnusamy
- PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (H.-C.C.); (V.K.P.)
- Department of Medicinal and Applied Chemistry & Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hua-Chen Chan
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
- Department of Medical Laboratory Science, College of Medicine, I-Shou University, Kaohsiung 824005, Taiwan
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
- Graduate Institute of Medicine, College of Medicine & Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
- Center for Lipid Biosciences, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
| |
Collapse
|
87
|
Zhu LR, He XH, Yuan YH, Yuan H, Xia XH. [Changes and significance of oxidized phospholipids and endothelial nitric oxide synthase in the acute stage of Kawasaki disease]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:829-834. [PMID: 39148387 PMCID: PMC11334547 DOI: 10.7499/j.issn.1008-8830.2403056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/04/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVES To investigate the changes in the serum levels of oxidized phospholipids (OxPLs) and endothelial nitric oxide synthase (eNOS) and their association with coronary artery disease (CAL) in children in the acute stage of Kawasaki disease (KD), as well as the clinical significance of OxPLs and eNOS. METHODS A prospective study was conducted on 95 children in the acute stage of KD (KD group). According to the presence of absence of CAL, the KD group was further divided into a CAL subgroup and a non-CAL (NCAL) subgroup. Thirty children with fever due to lower respiratory tract infection were enrolled as the fever group. Thirty healthy children who underwent physical examination were enrolled as the healthy control group. The above groups were compared in terms of general information and serum levels of OxPLs, eNOS and other laboratory indexes, and the correlation between OxPLs level and eNOS level was analyzed. RESULTS The KD group had a significantly higher level of OxPLs and a significantly lower level of eNOS compared with the fever group and the healthy control group (P<0.05). After treatment, the children with KD had a significantly decreased OxPLs level and a significantly increased eNOS level (P<0.05). Compared with the NCAL subgroup, the CAL subgroup had a significantly higher level of OxPLs and a significantly lower level of eNOS (P<0.05). Among the children of KD, the level of OxPLs was negatively correlated with that of eNOS (rs=-0.353, P<0.05). CONCLUSIONS Serum OxPLs and eNOS in the acute stage of KD may be involved in the development of CAL in children with KD, and therefore, they may be used as the biomarkers to predict CAL in these children.
Collapse
Affiliation(s)
- Liu-Rong Zhu
- Department of Pediatrics, First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Xue-Hua He
- Department of Pediatrics, First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Yong-Hua Yuan
- Department of Pediatrics, First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | | | | |
Collapse
|
88
|
Zhang Y, Gong J, Hu X, He L, Lin Y, Zhang J, Meng X, Zhang Y, Mo J, Day DB, Xiang J. Glycerophospholipid metabolism changes association with ozone exposure. JOURNAL OF HAZARDOUS MATERIALS 2024; 475:134870. [PMID: 38876019 DOI: 10.1016/j.jhazmat.2024.134870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 06/16/2024]
Abstract
Exposure to ozone (O3) has been associated with cardiovascular outcomes in humans, yet the underlying mechanisms of the adverse effect remain poorly understood. We aimed to investigate the association between O3 exposure and glycerophospholipid metabolism in healthy young adults. We quantified plasma concentrations of phosphatidylcholines (PCs) and lysophosphatidylcholines (lysoPCs) using a UPLC-MS/MS system. Time-weighted personal exposures were calculated to O3 and co-pollutants over 4 time windows, and we employed orthogonal partial least squares discriminant analysis to discern differences in lipids profiles between high and low O3 exposure. Linear mixed-effects models and mediation analysis were utilized to estimate the associations between O3 exposure, lipids, and cardiovascular physiology indicators. Forty-three healthy adults were included in this study, and the mean (SD) time-weighted personal exposures to O3 was 9.08 (4.06) ppb. With shorter exposure durations, O3 increases were associated with increasing PC and lysoPC levels; whereas at longer exposure times, the opposite relationship was shown. Furthermore, two specific lipids, namely lysoPC a C26:0 and lysoPC a C17:0, showed significantly positive mediating effects on associations of long-term O3 exposure with pulse wave velocity and systolic blood pressure, respectively. Alterations in specific lipids may underlie the cardiovascular effects of O3 exposure.
Collapse
Affiliation(s)
- Yi Zhang
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, China
| | - Jicheng Gong
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, China.
| | - Xinyan Hu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, China
| | - Linchen He
- College of Health, Lehigh University, Bethlehem, PA 19019, United States; Global Health Institute, Nicholas School of the Environment, Duke University, Durham, NC 27708, United States
| | - Yan Lin
- Global Health Institute, Nicholas School of the Environment, Duke University, Durham, NC 27708, United States
| | - Junfeng Zhang
- Global Health Institute, Nicholas School of the Environment, Duke University, Durham, NC 27708, United States
| | - Xin Meng
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, China
| | - Yinping Zhang
- Department of Building Science, Tsinghua University, Beijing 100084, China
| | - Jinhan Mo
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
| | - Drew B Day
- Seattle Children's Research Institute, Seattle, WA 98121, United States
| | - Jianbang Xiang
- School of Public Health, Sun Yat-Sen University, Shenzhen 518107, China
| |
Collapse
|
89
|
Mathews IT, Saminathan P, Henglin M, Liu M, Nadig N, Fang C, Mercader K, Chee SJ, Campbell AM, Patel AA, Tiwari S, Watrous JD, Ramesh K, Dicker M, Dao K, Meyer MA, Jousilahti P, Havulinna AS, Niiranen T, Salomaa V, Joosten LA, Netea MG, Zheng P, Kronenberg M, Patel SP, Gutkind JS, Ottensmeier C, Long T, Kaech SM, Hedrick CC, Cheng S, Jain M, Sharma S. Linoleoyl-lysophosphatidylcholine suppresses immune-related adverse events due to immune checkpoint blockade. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24310974. [PMID: 39148854 PMCID: PMC11326322 DOI: 10.1101/2024.08.07.24310974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Immune related adverse events (irAEs) after immune checkpoint blockade (ICB) therapy occur in a significant proportion of cancer patients. To date, the circulating mediators of ICB-irAEs remain poorly understood. Using non-targeted mass spectrometry, here we identify the circulating bio-active lipid linoleoyl-lysophosphatidylcholine (LPC 18:2) as a modulator of ICB-irAEs. In three independent human studies of ICB treatment for solid tumor, loss of circulating LPC 18:2 preceded the development of severe irAEs across multiple organ systems. In both healthy humans and severe ICB-irAE patients, low LPC 18:2 was found to correlate with high blood neutrophilia. Reduced LPC 18:2 biosynthesis was confirmed in preclinical ICB-irAE models, and LPC 18:2 supplementation in vivo suppressed neutrophilia and tissue inflammation without impacting ICB anti-tumor response. Results indicate that circulating LPC 18:2 suppresses human ICB-irAEs, and LPC 18:2 supplementation may improve ICB outcomes by preventing severe inflammation while maintaining anti-tumor immunity.
Collapse
Affiliation(s)
- Ian T. Mathews
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Department of Medicine, University of California San Diego, La Jolla CA 92093
| | | | - Mir Henglin
- Cedars Sinai Medical Center, Los Angeles CA 90048
| | - Mingyue Liu
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201
| | | | - Camille Fang
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Kysha Mercader
- Department of Medicine, University of California San Diego, La Jolla CA 92093
| | - Serena J. Chee
- University of Southampton, Southampton, United Kingdom
- Institute of Systems, Molecular and Integrative Biology,University of Liverpool, Liverpool, United Kingdom
| | | | | | - Saumya Tiwari
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Jeramie D. Watrous
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Karthik Ramesh
- Department of Medicine, University of California San Diego, La Jolla CA 92093
| | | | - Khoi Dao
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | | | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Aki S. Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland, FIMM-HiLIFE, Helsinki, Finland
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Internal Medicine, University of Turku, Turku, Finland
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Genomics and Immunometabolism, Life and Medical Sciences Institute, University of Bonn, Germany
| | - Pan Zheng
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Department of Molecular Biology, University of California San Diego, La Jolla CA 92093
| | - Sandip Pravin Patel
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla CA 92037
| | - J. Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla CA 92037
- Department of Pharmacology, University of California San Diego, La Jolla CA 92093
| | - Christian Ottensmeier
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Institute of Systems, Molecular and Integrative Biology,University of Liverpool, Liverpool, United Kingdom
| | - Tao Long
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Susan M. Kaech
- Salk Institute for Biological Studies, La Jolla CA 92037
| | - Catherine C. Hedrick
- Immunology Center of Georgia and Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Susan Cheng
- Cedars Sinai Medical Center, Los Angeles CA 90048
| | - Mohit Jain
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Sonia Sharma
- La Jolla Institute for Immunology, La Jolla, CA 92037
| |
Collapse
|
90
|
Maekawa M, Iwahori A, Kumondai M, Sato Y, Sato T, Mano N. Determination of Choline-Containing Compounds in Rice Bran Fermented with Aspergillus oryzae Using Liquid Chromatography/Tandem Mass Spectrometry. Mass Spectrom (Tokyo) 2024; 13:A0151. [PMID: 39161737 PMCID: PMC11331278 DOI: 10.5702/massspectrometry.a0151] [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: 05/13/2024] [Accepted: 07/19/2024] [Indexed: 08/21/2024] Open
Abstract
Choline-containing compounds are essential nutrients for human activity, as they are involved in many biological processes, including cell membrane organization, methyl group donation, neurotransmission, signal transduction, lipid transport, and metabolism. These compounds are normally obtained from food. Fermented brown rice and rice bran with Aspergillus oryzae (FBRA) is a fermented food product derived from rice and rice ingredients. FBRA exhibits a multitude of functional properties with respect to the health sciences. This study has a particular focus on choline-containing compounds. We first developed a simultaneous liquid chromatography/tandem mass spectrometry (LC/MS/MS) analysis method for seven choline-containing compounds. The method was subsequently applied to FBRA and its ingredients. Hydrophilic interaction chromatography (HILIC) and selected reaction monitoring were employed for the simultaneous analysis of seven choline-containing compounds. MS ion source conditions were optimized in positive ion mode, and the product ions derived from the choline group were obtained through MS/MS optimization. Under optimized HILIC conditions, the peaks exhibited good shape without peak tailing. Calibration curves demonstrated high linearity across a 300- to 10,000-fold concentration range. The application of the method to FBRA and other ingredients revealed significant differences between food with and without fermentation. In particular, betaine and α-glycerophosphocholine were found to be highest in FBRA and brown rice malt, respectively. The results indicated that the fermentation processing of rice ingredients results in alterations to the choline-containing compounds present in foods. The developed HILIC/MS/MS method proved to be a valuable tool for elucidating the composition of choline-containing compounds in foods.
Collapse
Affiliation(s)
- Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1–1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980–8574, Japan
- Faculty of Pharmaceutical Sciences, Tohoku University, 1–1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980–8574, Japan
| | - Anna Iwahori
- Faculty of Pharmaceutical Sciences, Tohoku University, 1–1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980–8574, Japan
| | - Masaki Kumondai
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1–1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980–8574, Japan
| | - Yu Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1–1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980–8574, Japan
| | - Toshihiro Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1–1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980–8574, Japan
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1–1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980–8574, Japan
- Faculty of Pharmaceutical Sciences, Tohoku University, 1–1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980–8574, Japan
| |
Collapse
|
91
|
Mu W, Han X, Tong M, Ben S, Yao M, Zhao Y, Xia J, Ren L, Huang C, Li D, Li X, Jiang Q, Yan B. Identification of the Metabolic Signature of Aging Retina. Transl Vis Sci Technol 2024; 13:8. [PMID: 39102240 PMCID: PMC11309042 DOI: 10.1167/tvst.13.8.8] [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: 03/12/2024] [Accepted: 06/26/2024] [Indexed: 08/06/2024] Open
Abstract
Purpose This study aims to explore the metabolic signature of aging retina and identify the potential metabolic biomarkers for the diagnosis of retinal aging. Methods Retinal samples were collected from both young (two months) and aging (14 months) mice to conduct an unbiased metabolic profiling. Liquid chromatography-tandem mass spectrometry analysis was conducted to screen for the metabolic biomarkers and altered signaling pathways associated with retinal aging. Results We identified 166 metabolites differentially expressed between young and aged retinas using a threshold of orthogonal projection to latent structures discriminant analysis variable importance in projection >1 and P < 0.05. These metabolites were significantly enriched in several metabolic pathways, including purine metabolism, citrate cycle, phenylalanine, tyrosine and tryptophan biosynthesis, glycerophospholipid metabolism, and alanine, aspartate and glutamate metabolism. Among these significantly enriched pathways, glycerophospholipid metabolites emerged as promising candidates for retinal aging biomarkers. We assessed the potential of these metabolites as biomarkers through an analysis of their sensitivity and specificity, determined by the area under the receiver-operating characteristic (ROC) curves. Notably, the metabolites like PC (15:0/22:6), PC (17:0/14:1), LPC (P-16:0), PE (16:0/20:4), and PS (17:0/16:1) demonstrated superior performance in sensitivity, specificity, and accuracy in predicting retinal aging. Conclusions This study sheds light on the molecular mechanisms underlying retinal aging by identifying distinct metabolic profiles and pathways. These findings provide a valuable foundation for developing future clinical applications in diagnosing, identifying, and treating age-related retinal degeneration. Translational Relevance This study sheds light on novel metabolic profiles and biomarkers in aging retinas, potentially paving the way for targeted interventions in preventing, diagnosing, and treating age-related retinal degeneration and other retinal diseases.
Collapse
Affiliation(s)
- Wan Mu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Han
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Ming Tong
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Shuai Ben
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mudi Yao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiao Xia
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Ren
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Chang Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Duo Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Xiumiao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
92
|
Wang X, Zhao B, Ruan Y, Xu W, Luo Z, Xu J, Shi C, Shan J. Fagopyrum Dibotrys Rhizoma regulates pulmonary lipid metabolic homeostasis and the ERK-cPLA 2 pathway to alleviate asthma in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155782. [PMID: 38851102 DOI: 10.1016/j.phymed.2024.155782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/04/2024] [Accepted: 05/26/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Asthma is a complex disease with mechanisms involving multiple factors, and there is still a lack of highly effective and low-side-effect drugs. Traditional Chinese medicine Fagopyrum Dibotrys Rhizoma (FDR) has been applied for the treatment of acute and chronic bronchitis as well as bronchial asthma due to its favorable pharmacological activity. However, the exact mechanism of FDR remains unclear. OBJECTIVE A mouse model of asthma was created using OVA and HDM. To investigate the mechanism of FDR in asthma treatment, a combination of network pharmacology, lipidomics, and molecular biology approaches was employed. METHODS To evaluate the therapeutic effects of FDR on asthma, we established two distinct models of asthma in C57BL/6 J mice using OVA and HDM, respectively. We then employed LC-MS to analyze the major chemical constituents in FDR. Next, the network pharmacology approach was used to predict the potential targets and mechanisms of FDR in asthma treatment. Additionally, lipidomics analysis of mouse serum was conducted using LC-MS. Finally, the impact of FDR on the ERK -cPLA2 signaling pathway was investigated through Western Blotting assay. RESULTS FDR treatment has been shown to improve histomorphological changes, lung function and inflammation in models of OVA and HDM-induced asthma. Using UPLC/LTQ-Orbitrap-MS, we were able to identify 12 potential active components. Network pharmacology analysis revealed that FDR shares 75 targets with asthma. Further analysis using GO and KEGG pathways demonstrated the involvement of key pathways such as PI3K-Akt, TNF, and MAPK. Additionally, lipidomics analysis of the serum from OVA and HDM induced asthma mice showed disturbances in lipid metabolism, which were effectively ameliorated by FDR treatment. Mechanistically, FDR inhibits ERK1/2-cPLA2, leading to a reduction in lysophospholipids and restoration of lipid balance, thereby aiding in the treatment of asthma. CONCLUSION FDR has been shown to improve lipid metabolism disorder in the serum of asthmatic mice, thereby potentially serving as a treatment for asthma. This can be achieved by regulating the activation levels of ERK1/2 and p38MAPK. Consequently, the production of lysophosphatide is reduced, thereby alleviating the disorder of lipid metabolism and achieving the desired therapeutic effect in asthma treatment.
Collapse
Affiliation(s)
- Xuan Wang
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Binshu Zhao
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuyuan Ruan
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weichen Xu
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zichen Luo
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jianya Xu
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Shi
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jinjun Shan
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
93
|
Aghaei M, Talari FS, Mollahosseini A, Keramati M. Validation of a high-performance liquid chromatography method for determining lysophosphatidylcholine content in bovine pulmonary surfactant medication. Biomed Chromatogr 2024; 38:e5926. [PMID: 38881378 DOI: 10.1002/bmc.5926] [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: 01/24/2024] [Revised: 05/08/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024]
Abstract
Pulmonary surfactant replacement therapy is a promising improvement in neonatal care for infants with respiratory distress syndrome. Lysophosphatidylcholine (LPC) is an undesirable component that can hinder surfactant proteins from enhancing the adsorption of surfactant lipids to balance surface tensions by creating a saturated coating on the interior of the lungs. A novel normal-phase liquid chromatography method utilizing UV detection and non-toxic solvents was developed and validated for the first time to analyze LPC in the complex matrix of pulmonary surfactant medication. The analytical method validation included evaluation of system suitability, repeatability, intermediate precision, linearity, accuracy, limit of detection (LOD), limit of quantification (LOQ), stability and robustness. The method yielded detection and quantification limits of 4.4 and 14.5 μg/ml, respectively. The calibration curve was modified linearly within the LOQ to 1.44 mg/ml range, with a determination coefficient of 0.9999 for standards and 0.9997 for sample solutions. Given the lack of reliable published data on LPC analysis in pulmonary surfactant medications, this newly developed method demonstrates promising results and offers advantages of HPLC methodology, including simplicity, accuracy, specificity, sensitivity and an exceptionally low LOD and LOQ. These attributes contribute to considering this achievement as an innovative method.
Collapse
Affiliation(s)
- Mahsa Aghaei
- ARC Bioassay (Iran Food and Drug Administration Accredited QC Laboratory of Biopharmaceutical Products), Tehran, Iran
- Department of Chemistry, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Faezeh Shirgaei Talari
- ARC Bioassay (Iran Food and Drug Administration Accredited QC Laboratory of Biopharmaceutical Products), Tehran, Iran
| | - Afsaneh Mollahosseini
- Research Laboratory of Spectrometry & Micro and Nano Extraction, Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Malihe Keramati
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
94
|
Zhang H, Zhang S, Chen L, Xu R, Zhu J. LC-HRMS-based metabolomics and lipidomics analyses of a novel probiotic Akkermansia Muciniphila in response to different nutritional stimulations. J Microbiol Methods 2024; 223:106975. [PMID: 38889842 DOI: 10.1016/j.mimet.2024.106975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
The mucin-degrading gut commensal Akkermansia muciniphila (A. muciniphila) negatively correlates with various diseases, including metabolic disorders, neurodegenerative disorders, and cancers, through interacting with host receptors by diverse molecules. Still, their exact metabolic capability within the nutrient-rich environment (such as in the human gut) is not fully characterized. Therefore, in the present study, we investigated the comprehensive metabolome and lipidome of A. muciniphila after supplementation of four major gut microbial nutrients: mucin, inorganic salts, bile salts, and short-chain fatty acids (SCFAs). Our results showed that mucin is the predominant driver of the different lipidomic and metabolomic profiles of A. muciniphila, and it promotes the overall growth of this bacteria. While the addition of inorganic salts, bile salts, and SCFAs was found to inhibit the growth of A. muciniphila. Interestingly, inorganic salts affected the purine metabolism in A. muciniphila cultures, while adding bile salts significantly increased the production of other bile acids and N-acyl amides. Lastly, SCFAs were identified to alter the A. muciniphila energy utilization of triglycerides, fatty acyls, and phosphatidylethanolamines. To our knowledge, this is the first study to examine the comprehensive lipidome and metabolome of A. muciniphila, which highlights the importance of nutritional impacts on the lipidome and metabolome of A. muciniphila and hence providing foundational knowledge to unveil the potential effects of A. muciniphila on host health.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Human Sciences & James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States of America
| | - Shiqi Zhang
- Department of Human Sciences & James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States of America
| | - Li Chen
- Department of Human Sciences & James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States of America
| | - Rui Xu
- Department of Human Sciences & James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States of America
| | - Jiangjiang Zhu
- Department of Human Sciences & James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States of America.
| |
Collapse
|
95
|
Arbeev KG, Bagley O, Ukraintseva SV, Kulminski A, Stallard E, Schwaiger-Haber M, Patti GJ, Gu Y, Yashin AI, Province MA. Methods for joint modelling of longitudinal omics data and time-to-event outcomes: Applications to lysophosphatidylcholines in connection to aging and mortality in the Long Life Family Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.29.24311176. [PMID: 39132492 PMCID: PMC11312646 DOI: 10.1101/2024.07.29.24311176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Studying relationships between longitudinal changes in omics variables and risks of events requires specific methodologies for joint analyses of longitudinal and time-to-event outcomes. We applied two such approaches (joint models [JM], stochastic process models [SPM]) to longitudinal metabolomics data from the Long Life Family Study focusing on understudied associations of longitudinal changes in lysophosphatidylcholines (LPC) with mortality and aging-related outcomes (23 LPC species, 5,790 measurements of each in 4,011 participants, 1,431 of whom died during follow-up). JM analyses found that higher levels of the majority of LPC species were associated with lower mortality risks, with the largest effect size observed for LPC 15:0/0:0 (hazard ratio: 0.715, 95% CI (0.649, 0.788)). SPM applications to LPC 15:0/0:0 revealed how the association found in JM reflects underlying aging-related processes: decline in robustness to deviations from optimal LPC levels, better ability of males' organisms to return to equilibrium LPC levels (which are higher in females), and increasing gaps between the optimum and equilibrium levels leading to increased mortality risks with age. Our results support LPC as a biomarker of aging and related decline in robustness/resilience, and call for further exploration of factors underlying age-dynamics of LPC in relation to mortality and diseases.
Collapse
Affiliation(s)
- Konstantin G. Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Svetlana V. Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Alexander Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Michaela Schwaiger-Haber
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Center for Metabolomics and Isotope Tracing at Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Gary J. Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Center for Metabolomics and Isotope Tracing at Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Yian Gu
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York 10032, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Anatoliy I. Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Michael A. Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
96
|
Oft HC, Simon DW, Sun D. New insights into metabolism dysregulation after TBI. J Neuroinflammation 2024; 21:184. [PMID: 39075578 PMCID: PMC11288120 DOI: 10.1186/s12974-024-03177-6] [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: 05/01/2024] [Accepted: 07/16/2024] [Indexed: 07/31/2024] Open
Abstract
Traumatic brain injury (TBI) remains a leading cause of death and disability that places a great physical, social, and financial burden on individuals and the health system. In this review, we summarize new research into the metabolic changes described in clinical TBI trials, some of which have already shown promise for informing injury classification and staging. We focus our discussion on derangements in glucose metabolism, cell respiration/mitochondrial function and changes to ketone and lipid metabolism/oxidation to emphasize potentially novel biomarkers for clinical outcome prediction and intervention and offer new insights into possible underlying mechanisms from preclinical research of TBI pathology. Finally, we discuss nutrition supplementation studies that aim to harness the gut/microbiome-brain connection and manipulate systemic/cellular metabolism to improve post-TBI recovery. Taken together, this narrative review summarizes published TBI-associated changes in glucose and lipid metabolism, highlighting potential metabolite biomarkers for clinical use, the cellular processes linking these markers to TBI pathology as well as the limitations and future considerations for TBI "omics" work.
Collapse
Affiliation(s)
- Helena C Oft
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dennis W Simon
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
97
|
Hou J, Wang X, Zhang J, Shen Z, Li X, Yang Y. Chuanxiong Renshen Decoction Inhibits Alzheimer's Disease Neuroinflammation by Regulating PPARγ/NF-κB Pathway. Drug Des Devel Ther 2024; 18:3209-3232. [PMID: 39071817 PMCID: PMC11283787 DOI: 10.2147/dddt.s462266] [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: 04/22/2024] [Accepted: 07/12/2024] [Indexed: 07/30/2024] Open
Abstract
Background and Aim Previous studies of our research group have shown that Chuanxiong Renshen Decoction (CRD) has the effect of treating AD, but the exact mechanism of its effect is still not clarified. The aim of this study was to investigate the effect and mechanism of CRD on AD neuroinflammation. Materials and Methods Morris Water Maze (MWM) tests were employed to assess the memory and learning capacity of AD mice. HE and Nissl staining were used to observe the neural cells of mice. The expression of Iba-1 and CD86 were detected by immunohistochemical staining. Utilize UHPLC-MS/MS metabolomics techniques and the KEGG to analyze the metabolic pathways of CRD against AD. Lipopolysaccharide (LPS) induced BV2 microglia cells to construct a neuroinflammatory model. The expression of Iba-1 and CD86 were detected by immunofluorescence and flow cytometry. The contents of TNF-α and IL-1β were detected by ELISA. Western blot assay was used to detect the expression of PPARγ, p-NF-κB p65, NF-κB p65 proteins and inflammatory cytokines iNOS and COX-2 in PPARγ/NF-κB pathway with and without PPARγ inhibitor GW9662. Results CRD ameliorated the learning and memory ability of 3×Tg-AD mice, repaired the damaged nerve cells in the hippocampus, reduced the area of Iba-1 and CD86 positive areas in both the hippocampus and cortex regions, as well as attenuated serum levels of IL-1β and TNF-α in mice. CRD-containing serum significantly decreased the expression level of Iba-1, significantly reduced the levels of TNF-α and IL-1β, significantly increased the protein expression of PPARγ, and significantly decreased the proteins expression of iNOS, COX-2 and p-NF-κB p65 in BV2 microglia cells. After addition of PPARγ inhibitor GW9662, the inhibitory effect of CRD-containing serum on NF-κB activation was significantly weakened. Conclusion CRD can activate PPARγ, regulating PPARγ/NF-κB signaling pathway, inhibiting microglia over-activation and reducing AD neuroinflammation.
Collapse
Affiliation(s)
- Jinling Hou
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Xiaoyan Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Jian Zhang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Zhuojun Shen
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Xiang Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Yuanxiao Yang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, People’s Republic of China
| |
Collapse
|
98
|
Lu W, Li Y, Ge L, Wang H, Liu T, Zhao Q, Mao Z, Liang J, Wang P, Chen K, Xue J, Shen Q. Comprehensive lipidomics study of basa catfish and sole fish using ultra-performance liquid chromatography Q-extractive orbitrap mass spectrometry for fish authenticity. Curr Res Food Sci 2024; 9:100812. [PMID: 39139808 PMCID: PMC11321432 DOI: 10.1016/j.crfs.2024.100812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/23/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
The authenticity of fish products has become a widespread issue in markets due to substitution and false labeling. Lipidomics combined with chemometrics enables the fraudulence identification of food through the analysis of a large amount of data. This study utilized ultra-high-performance liquid chromatography (UHPLC)-QE Orbitrap MS technology to comprehensively analyze the lipidomics of commercially available basa catfish and sole fish. In positive and negative ion modes, a total of 779 lipid molecules from 21 lipid subclasses were detected, with phospholipid molecules being the most abundant, followed by glycerides molecules. Significant differences in the lipidome fingerprinting between the two fish species were observed. A total of 165 lipid molecules were screened out as discriminative features to distinguish between basa catfish and sole fish, such as TAG(16:0/16:0/18:1), PC(14:0/22:3), and TAG(16:1/18:1/18:1), etc. This study could provide valuable insights into authenticating aquatic products through comprehensive lipidomics analysis, contributing to quality control and consumer protection in the food industry.
Collapse
Affiliation(s)
- Weibo Lu
- Collaborative Innovation Center of Seafood Deep Processing, Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Yunyan Li
- Collaborative Innovation Center of Seafood Deep Processing, Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Lijun Ge
- Collaborative Innovation Center of Seafood Deep Processing, Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Honghai Wang
- Collaborative Innovation Center of Seafood Deep Processing, Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Ting Liu
- Zhoushan Institute of Food & Drug Control, Zhoushan, China
| | - Qiaoling Zhao
- Zhoushan Institute of Food & Drug Control, Zhoushan, China
| | - Zhujun Mao
- Panvascular Diseases Research Center, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Jingjing Liang
- Zhejiang Provincial Institute for Food and Drug Control, Hangzhou, 310052, China
| | - Pingya Wang
- Zhoushan Institute of Food & Drug Control, Zhoushan, China
| | - Kang Chen
- Collaborative Innovation Center of Seafood Deep Processing, Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Jing Xue
- Collaborative Innovation Center of Seafood Deep Processing, Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Qing Shen
- Panvascular Diseases Research Center, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
- Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China
| |
Collapse
|
99
|
Zhou Q, Chang C, Wang Y, Gai X, Chen Y, Gao X, Liang Y, Sun Y. Comparative analysis of lysophospholipid metabolism profiles and clinical characteristics in patients with high vs. low C-reactive protein levels in acute exacerbations of chronic obstructive pulmonary disease. Clin Chim Acta 2024; 561:119816. [PMID: 38885755 DOI: 10.1016/j.cca.2024.119816] [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: 03/27/2024] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND The precise role of lysophospholipids (LysoPLs) in the pathogenesis of acute exacerbations of Chronic Obstructive Pulmonary Disease (AECOPD) remains unclear. In this study, we sought to elucidate the differences in serum LysoPL metabolite profiles and their correlation with clinical features between patients with low versus high CRP levels. METHODS A total of 58 patients with AECOPD were enrolled in the study. Patients were classified into two groups: low CRP group (CRP < 20 mg/L, n = 34) and high CRP group (CRP ≥ 20 mg/L, n = 24). Clinical data were collected, and the LysoPL metabolite profiles were analyzed using Liquid Chromatography-Mass Spectrometry (LC-MS) and identified by matching with the LipidBlast library. RESULTS Nineteen differential LysoPLs were initially identified through Student's t-test (p < 0.05 and VIP > 1). Subsequently, four LysoPLs, LPC(16:0), LPE(18:2), LPC(22:0), and LPC(24:0), were identified by FDR adjustment (adjusted p < 0.05). These four lysoPLs had a significant negative correlation with CRP. Integrative analysis revealed that LPC (16:0) and LPC (22:0) correlated with less hypercapnic respiratory failure and ICU admission. CONCLUSION AECOPD patients with high CRP levels demonstrated a distinctive LysoPL metabolism profile, with LPC (16:0), LPE(18:2), LPC(22:0), and LPC(24:0) being the most significantly altered lipid molecules. These alterations were associated with poorer clinical outcomes.
Collapse
Affiliation(s)
- Qiqiang Zhou
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Chun Chang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China; Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing, China
| | - Yating Wang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China; Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing, China
| | - Yahong Chen
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China; Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing, China
| | - Xu Gao
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Ying Liang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China; Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing, China.
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China; Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing, China
| |
Collapse
|
100
|
Fernandes T, Melo T, Conde T, Neves B, Domingues P, Resende R, Pereira CF, Moreira PI, Domingues MR. Mapping the lipidome in mitochondria-associated membranes (MAMs) in an in vitro model of Alzheimer's disease. J Neurochem 2024; 168:1237-1253. [PMID: 38327008 DOI: 10.1111/jnc.16072] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/06/2023] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
The disruption of mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) plays a relevant role in Alzheimer's disease (AD). MAMs have been implicated in neuronal dysfunction and death since it is associated with impairment of functions regulated in this subcellular domain, including lipid synthesis and trafficking, mitochondria dysfunction, ER stress-induced unfolded protein response (UPR), apoptosis, and inflammation. Since MAMs play an important role in lipid metabolism, in this study we characterized and investigated the lipidome alterations at MAMs in comparison with other subcellular fractions, namely microsomes and mitochondria, using an in vitro model of AD, namely the mouse neuroblastoma cell line (N2A) over-expressing the APP familial Swedish mutation (APPswe) and the respective control (WT) cells. Phospholipids (PLs) and fatty acids (FAs) were isolated from the different subcellular fractions and analyzed by HILIC-LC-MS/MS and GC-MS, respectively. In this in vitro AD model, we observed a down-regulation in relative abundance of some phosphatidylcholine (PC), lysophosphatidylcholine (LPC), and lysophosphatidylethanolamine (LPE) species with PUFA and few PC with saturated and long-chain FA. We also found an up-regulation of CL, and antioxidant alkyl acyl PL. Moreover, multivariate analysis indicated that each organelle has a specific lipid profile adaptation in N2A APPswe cells. In the FAs profile, we found an up-regulation of C16:0 in all subcellular fractions, a decrease of C18:0 levels in total fraction (TF) and microsomes fraction, and a down-regulation of 9-C18:1 was also found in mitochondria fraction in the AD model. Together, these results suggest that the over-expression of the familial APP Swedish mutation affects lipid homeostasis in MAMs and other subcellular fractions and supports the important role of lipids in AD physiopathology.
Collapse
Affiliation(s)
- Tânia Fernandes
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Tânia Melo
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Tiago Conde
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Bruna Neves
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Rosa Resende
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Cláudia F Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria Rosário Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| |
Collapse
|