1
|
Ström A, Stenlund H, Ohlsson B. The Metabolomic Profile of Microscopic Colitis Is Affected by Smoking but Not Histopathological Diagnosis, Clinical Course, Symptoms, or Treatment. Metabolites 2024; 14:303. [PMID: 38921438 PMCID: PMC11205623 DOI: 10.3390/metabo14060303] [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: 04/30/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Microscopic colitis (MC) is classified as collagenous colitis (CC) and lymphocytic colitis (LC). Genetic associations between CC and human leucocyte antigens (HLAs) have been found, with smoking being a predisposing external factor. Smoking has a great impact on metabolomics. The aim of this explorative study was to analyze global metabolomics in MC and to examine whether the metabolomic profile differed regarding the type and course of MC, the presence of IBS-like symptoms, treatment, and smoking habits. Of the 240 identified women with MC aged ≤73 years, 131 completed the study questionnaire; the Rome III questionnaire; and the Visual Analog Scale for Irritable Bowel Syndrome (VAS-IBS). Blood samples were analyzed by ultra-high-performance liquid chromatograph mass spectrometry (UHLC-MS/UHPLC-MSMS). The women, 63.1 (58.7-67.2) years old, were categorized based on CC (n = 76) and LC (n = 55); one episode or refractory MC; IBS-like symptoms or not; use of corticosteroids or not; and smoking habits. The only metabolomic differences found in the univariate model after adjustment for false discovery rate (FDR) were between smokers and non-smokers. Serotonin was markedly increased in smokers (p < 0.001). No clear patterns appeared when conducting a principal component analysis (PCA). No differences in the metabolomic profile were found depending on the type or clinical course of the disease, neither in the whole MC group nor in the subgroup analysis of CC.
Collapse
Affiliation(s)
- Axel Ström
- Clinical Studies Sweden—Forum South, Skåne University Hospital, 22185 Lund, Sweden;
| | - Hans Stenlund
- Umeå Plant Science Centre (UPSC), Department of Plant Physiology, Umeå University, 90187 Umeå, Sweden;
| | - Bodil Ohlsson
- Department of Clinical Scineces, Lund University, 22100 Lund, Sweden
- Department of Internal Medicine, Skane University Hospital, 20502 Malmö, Sweden
| |
Collapse
|
2
|
Roth B, Ohlsson B. Microscopic colitis found together with celiac disease in a female population is associated with one episode of lymphocytic colitis. BMC Gastroenterol 2024; 24:70. [PMID: 38347511 PMCID: PMC10860280 DOI: 10.1186/s12876-024-03158-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 02/04/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Microscopic colitis (MC) is considered a chronic disease associated with autoimmune disease, smoking, and drugs. The aim was to examine the association between MC and celiac disease, adjusted for smoking, considering subtypes and clinical course of the disease in a retrospectively collected female cohort. METHODS Women (n = 240), ≤ 73 years, diagnosed as MC in medical records or pathological registers were invited. One hundred and fifty-eight women accepted to be included. Participants completed a study questionnaire about sociodemographic factors, lifestyle habits, and medical history; the Rome III questionnaire; and the visual analog scale for irritable bowel syndrome (VAS-IBS). Participants were categorized into collagenous colitis (CC) (n = 92) and lymphocytic colitis (LC) (n = 66) or MC with one episode of the disease (n = 70) and refractory MC (n = 88). Presence of IBS-like symptoms were noted. Blood samples were collected and analyzed for anti-transglutaminase antibodies. Differences between groups were calculated and logistic regression was adjusted for smoking habits. RESULTS MC and celiac disease debuted simultaneously in half of the cases. Celiac disease was most prevalent in LC (12.1% vs. 3.3%; p = 0.05) and MC with one episode (12.9% vs. 2.3%; p = 0.01). Anti-transglutaminase antibodies were found in one patient with one episode of MC. Corticosteroid use was most often found in CC (37.0% vs. 21.2%; p = 0.037) and refractory MC (38.6% vs. 20.0%; p = 0.015). Past smokers were most prevalent in patients with one episode of MC (54.3 vs. 29.5%; p = 0.007). Current smoking was the smoking habit with highest prevalence of IBS-like symptoms. When adjusted for smoking habits, celiac disease was associated with LC (OR: 4.222; 95% CI: 1.020-17.469; p = 0.047) and tended to be inversely associated with refractory MC (OR: 0.210; 95% CI: 0.042-1.506; p = 0.058). CONCLUSION Celiac disease is most common in patients with one episode of LC. The question remains whether LC in combination with celiac disease should be classified as celiac disease or two different entities.
Collapse
Affiliation(s)
- Bodil Roth
- Department of Internal Medicine, Lund University, Skåne University Hospital, Jan Waldenströms Street 15, Floor 5, 205 02, Malmö, Sweden
| | - Bodil Ohlsson
- Department of Internal Medicine, Lund University, Skåne University Hospital, Jan Waldenströms Street 15, Floor 5, 205 02, Malmö, Sweden.
| |
Collapse
|
3
|
Hill EB, Tang M, Long JM, Kemp JF, Westcott JL, Hendricks AE, Reisdorph NA, Campbell WW, Krebs NF. mini-MED: study protocol for a randomized, multi-intervention, semi-controlled feeding trial of a Mediterranean-amplified vs. habitual Western dietary pattern for the evaluation of food-specific compounds and cardiometabolic health. Trials 2024; 25:101. [PMID: 38302990 PMCID: PMC10835998 DOI: 10.1186/s13063-024-07939-8] [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: 12/07/2023] [Accepted: 01/16/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Diet is among the most influential lifestyle factors impacting chronic disease risk. Nutrimetabolomics, the application of metabolomics to nutrition research, allows for the detection of food-specific compounds (FSCs) that can be used to connect dietary patterns, such as a Mediterranean-style (MED) diet, to health. This validation study is based upon analyses from a controlled feeding MED intervention, where our team identified FSCs from eight foods that can be detected in biospecimens after consumption and may therefore serve as food intake biomarkers. METHODS Individuals with overweight/obesity who do not habitually consume a MED dietary pattern will complete a 16-week randomized, multi-intervention, semi-controlled feeding study of isocaloric dietary interventions: (1) MED-amplified dietary pattern, containing 500 kcal/day from eight MED target foods: avocado, basil, cherry, chickpea, oat, red bell pepper, walnut, and a protein source (alternating between salmon or unprocessed, lean beef), and (2) habitual/Western dietary pattern, containing 500 kcal/day from six non-MED target foods: cheesecake, chocolate frozen yogurt, refined grain bread, sour cream, white potato, and unprocessed, lean beef. After a 2-week washout, participants complete four, 4-week intervention periods, with biospecimen sampling and outcome assessments at baseline and at intervention weeks 4, 8, 12, and 16. The primary outcome is change in the relative abundance of FSCs from the eight MED target foods in participant biospecimens from baseline to the end of each intervention period. Secondary outcomes include mean change in cardiometabolic health indicators, inflammatory markers, and adipokines. Exploratory outcomes include change in diversity and community composition of the gut microbiota. DISCUSSION Our stepwise strategy, beginning with identification of FSCs in whole diets and biospecimens, followed by relating these to health indicators will lead to improved methodology for assessment of dietary patterns and a better understanding of the relationship between food and health. This study will serve as a first step toward validating candidate food intake biomarkers and allow for assessment of relationships with cardiometabolic health. The identification of food intake biomarkers is critical to future research and has implications spanning health promotion and disease prevention for many chronic conditions. TRIAL REGISTRATION Registered at ClinicalTrials.gov: NCT05500976 ; Date of registration: August 15, 2022.
Collapse
Affiliation(s)
- Emily B Hill
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Minghua Tang
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Julie M Long
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jennifer F Kemp
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jamie L Westcott
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Audrey E Hendricks
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, 80204, USA
| | - Nichole A Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Wayne W Campbell
- Department of Nutrition Science, Purdue University, West Lafayette, IN, 47906, USA
| | - Nancy F Krebs
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
4
|
Hill EB, Reisdorph RM, Rasolofomanana-Rajery S, Michel C, Khajeh-Sharafabadi M, Doenges KA, Weaver N, Quinn K, Sutliff AK, Tang M, Borengasser SJ, Frank DN, O'Connor LE, Campbell WW, Krebs NF, Hendricks AE, Reisdorph NA. Salmon Food-Specific Compounds and Their Metabolites Increase in Human Plasma and Are Associated with Cardiometabolic Health Indicators Following a Mediterranean-Style Diet Intervention. J Nutr 2024; 154:26-40. [PMID: 37918675 PMCID: PMC10808825 DOI: 10.1016/j.tjnut.2023.10.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/25/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Nutrimetabolomics allows for the comprehensive analysis of foods and human biospecimens to identify biomarkers of intake and begin to probe their associations with health. Salmon contains hundreds of compounds that may provide cardiometabolic benefits. OBJECTIVES We used untargeted metabolomics to identify salmon food-specific compounds (FSCs) and their predicted metabolites that were found in plasma after a salmon-containing Mediterranean-style (MED) diet intervention. Associations between changes in salmon FSCs and changes in cardiometabolic health indicators (CHIs) were also explored. METHODS For this secondary analysis of a randomized, crossover, controlled feeding trial, 41 participants consumed MED diets with 2 servings of salmon per week for 2 5-wk periods. CHIs were assessed, and fasting plasma was collected pre- and postintervention. Plasma, salmon, and 99 MED foods were analyzed using liquid chromatography-mass spectrometry-based metabolomics. Compounds were characterized as salmon FSCs if detected in all salmon replicates but none of the other foods. Metabolites of salmon FSCs were predicted using machine learning. For salmon FSCs and metabolites found in plasma, linear mixed-effect models were used to assess change from pre- to postintervention and associations with changes in CHIs. RESULTS Relative to the other 99 MED foods, there were 508 salmon FSCs with 237 unique metabolites. A total of 143 salmon FSCs and 106 metabolites were detected in plasma. Forty-eight salmon FSCs and 30 metabolites increased after the intervention (false discovery rate <0.05). Increases in 2 annotated salmon FSCs and 2 metabolites were associated with improvements in CHIs, including total cholesterol, low-density lipoprotein cholesterol, triglycerides, and apolipoprotein B. CONCLUSIONS A data-driven nutrimetabolomics strategy identified salmon FSCs and their predicted metabolites that were detectable in plasma and changed after consumption of a salmon-containing MED diet. Findings support this approach for the discovery of compounds in foods that may serve, upon further validation, as biomarkers or act as bioactive components influential to health. The trials supporting this work were registered at NCT02573129 (Mediterranean-style diet intervention) and NCT05500976 (ongoing clinical trial).
Collapse
Affiliation(s)
- Emily B Hill
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Richard M Reisdorph
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sakaiza Rasolofomanana-Rajery
- Department of Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, United States; Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Cole Michel
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mobin Khajeh-Sharafabadi
- Department of Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, United States
| | - Katrina A Doenges
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Nicholas Weaver
- Department of Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, United States
| | - Kevin Quinn
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Aimee K Sutliff
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Minghua Tang
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sarah J Borengasser
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Daniel N Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lauren E O'Connor
- USDA, Agricultural Research Service, Beltsville Human Nutrition Research Center, Food Components and Health Laboratory, Beltsville, MD, United States
| | - Wayne W Campbell
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
| | - Nancy F Krebs
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Audrey E Hendricks
- Department of Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, United States; Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| | - Nichole A Reisdorph
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
5
|
Deng K, Gupta DK, Shu XO, Lipworth L, Zheng W, Thomas VE, Cai H, Cai Q, Wang TJ, Yu D. Metabolite Signature of Life's Essential 8 and Risk of Coronary Heart Disease Among Low-Income Black and White Americans. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:e004230. [PMID: 38014580 PMCID: PMC10843634 DOI: 10.1161/circgen.123.004230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/26/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Life's essential 8 (LE8) is a comprehensive construct of cardiovascular health. Yet, little is known about the LE8 score, its metabolic correlates, and their predictive implications among Black Americans and low-income individuals. METHODS In a nested case-control study of coronary heart disease (CHD) among 299 pairs of Black and 298 pairs of White low-income Americans from the Southern Community Cohort Study, we estimated LE8 score and applied untargeted plasma metabolomics and elastic net with leave-one-out cross-validation to identify metabolite signature (MetaSig) of LE8. Associations of LE8 score and MetaSig with incident CHD were examined using conditional logistic regression. The mediation effect of MetaSig on the LE8-CHD association was also examined. The external validity of MetaSig was evaluated in another nested CHD case-control study among 299 pairs of Chinese adults. RESULTS Higher LE8 score was associated with lower CHD risk (standardized odds ratio, 0.61 [95% CI, 0.53-0.69]). The MetaSig, consisting of 133 metabolites, showed significant correlation with LE8 score (r=0.61) and inverse association with CHD (odds ratio, 0.57 [0.49-0.65]), robust to adjustment for LE8 score and across participants with different sociodemographic and health status ([odds ratios, 0.42-0.69]; all P<0.05). MetaSig mediated a large portion of the LE8-CHD association: 53% (32%-80%). Significant associations of MetaSig with LE8 score and CHD risk were found in validation cohort (r=0.49; odds ratio, 0.57 [0.46-0.69]). CONCLUSIONS Higher LE8 score and its MetaSig were associated with lower CHD risk among low-income Black and White Americans. Metabolomics may offer an objective measure of LE8 and its metabolic phenotype relevant to CHD prevention among diverse populations.
Collapse
Affiliation(s)
- Kui Deng
- Vanderbilt Epidemiology Center and Division of Epidemiology, Dept of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Deepak K. Gupta
- Vanderbilt Translational & Clinical Cardiovascular Research Center & Division of Cardiovascular Medicine, Dept of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Xiao-Ou Shu
- Vanderbilt Epidemiology Center and Division of Epidemiology, Dept of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Loren Lipworth
- Vanderbilt Epidemiology Center and Division of Epidemiology, Dept of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Wei Zheng
- Vanderbilt Epidemiology Center and Division of Epidemiology, Dept of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Victoria E. Thomas
- Vanderbilt Translational & Clinical Cardiovascular Research Center & Division of Cardiovascular Medicine, Dept of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Hui Cai
- Vanderbilt Epidemiology Center and Division of Epidemiology, Dept of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Qiuyin Cai
- Vanderbilt Epidemiology Center and Division of Epidemiology, Dept of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Thomas J. Wang
- Dept of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Danxia Yu
- Vanderbilt Epidemiology Center and Division of Epidemiology, Dept of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
6
|
Salazar-Puerta AI, Rincon-Benavides MA, Cuellar-Gaviria TZ, Aldana J, Martinez GV, Ortega-Pineda L, Das D, Dodd D, Spencer CA, Deng B, McComb DW, Englert JA, Ghadiali S, Zepeda-Orozco D, Wold LE, Gallego-Perez D, Higuita-Castro N. Engineered Extracellular Vesicles Derived from Dermal Fibroblasts Attenuate Inflammation in a Murine Model of Acute Lung Injury. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210579. [PMID: 37119468 PMCID: PMC10573710 DOI: 10.1002/adma.202210579] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/29/2023] [Indexed: 06/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) represents a significant burden to the healthcare system, with ≈200 000 cases diagnosed annually in the USA. ARDS patients suffer from severe refractory hypoxemia, alveolar-capillary barrier dysfunction, impaired surfactant function, and abnormal upregulation of inflammatory pathways that lead to intensive care unit admission, prolonged hospitalization, and increased disability-adjusted life years. Currently, there is no cure or FDA-approved therapy for ARDS. This work describes the implementation of engineered extracellular vesicle (eEV)-based nanocarriers for targeted nonviral delivery of anti-inflammatory payloads to the inflamed/injured lung. The results show the ability of surfactant protein A (SPA)-functionalized IL-4- and IL-10-loaded eEVs to promote intrapulmonary retention and reduce inflammation, both in vitro and in vivo. Significant attenuation is observed in tissue damage, proinflammatory cytokine secretion, macrophage activation, influx of protein-rich fluid, and neutrophil infiltration into the alveolar space as early as 6 h post-eEVs treatment. Additionally, metabolomics analyses show that eEV treatment causes significant changes in the metabolic profile of inflamed lungs, driving the secretion of key anti-inflammatory metabolites. Altogether, these results establish the potential of eEVs derived from dermal fibroblasts to reduce inflammation, tissue damage, and the prevalence/progression of injury during ARDS via nonviral delivery of anti-inflammatory genes/transcripts.
Collapse
Affiliation(s)
- Ana I. Salazar-Puerta
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
| | - María A. Rincon-Benavides
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States
| | | | - Julian Aldana
- Biochemistry Program, The Ohio State University, Columbus, Ohio, United States
| | - Gabriela Vasquez Martinez
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States
| | - Lilibeth Ortega-Pineda
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Devleena Das
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Daniel Dodd
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biomedical Science Graduate Program, The Ohio State University, Columbus, Ohio, United States
| | - Charles A. Spencer
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| | - Binbin Deng
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, Ohio, United States
| | - David W. McComb
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, Ohio, United States
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Joshua A. Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Samir Ghadiali
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Diana Zepeda-Orozco
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States
- Division of Pediatric Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, Ohio, United States
| | - Loren E. Wold
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States
- Division of General Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States
- Division of General Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
7
|
Deng K, Gupta DK, Shu XO, Lipworth L, Zheng W, Thomas VE, Cai H, Cai Q, Wang TJ, Yu D. Metabolite Signature of Life's Essential 8 and Risk of Coronary Heart Disease among Low-Income Black and White Americans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.24.23289055. [PMID: 37163035 PMCID: PMC10168489 DOI: 10.1101/2023.04.24.23289055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Background and Aims Life's Essential 8 (LE8) is a comprehensive construct of cardiovascular health. Yet, little is known about LE8 score, its metabolic correlates, and their predictive implications among Black Americans and low-income individuals. Methods In a nested case-control study of coronary heart disease (CHD) among 598 Black and 596 White low-income Americans, we estimated LE8 score, conducted untargeted plasma metabolites profiling, and used elastic net with leave-one-out cross-validation to identify metabolite signature (MetaSig) of LE8. Associations of LE8 score and MetaSig with incident CHD were examined using conditional logistic regression. Mediation effect of MetaSig on the LE8-CHD association was also examined. The external validity of MetaSig was evaluated in another nested CHD case-control study among 598 Chinese adults. Results Higher LE8 score was associated with lower CHD risk [standardized OR (95% CI)=0.61 (0.53-0.69)]. The identified MetaSig, consisting of 133 metabolites, showed strong correlation with LE8 score ( r =0.61) and inverse association with CHD risk [OR (95% CI)=0.57 (0.49-0.65)], robust to adjustment for LE8 score and across participants with different sociodemographic and health status (ORs: 0.42-0.69; all P <0.05). MetaSig mediated a large portion of the LE8-CHD association: 53% (32%-80%) ( P <0.001). Significant associations of MetaSig with LE8 score and CHD risk were found in validation cohort [ r =0.49; OR (95% CI)=0.57 (0.46-0.69)]. Conclusions Higher LE8 score and its MetaSig were associated with lower CHD risk among low-income Black and White Americans. Metabolomics may offer an objective and comprehensive measure of LE8 score and its metabolic phenotype relevant to CHD prevention among diverse populations.
Collapse
|
8
|
Gea J, Enríquez-Rodríguez CJ, Pascual-Guardia S. Metabolomics in COPD. Arch Bronconeumol 2023; 59:311-321. [PMID: 36717301 DOI: 10.1016/j.arbres.2022.12.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/28/2022] [Accepted: 12/07/2022] [Indexed: 01/20/2023]
Abstract
The clinical presentation of chronic obstructive pulmonary disease (COPD) is highly heterogeneous. Attempts have been made to define subpopulations of patients who share clinical characteristics (phenotypes and treatable traits) and/or biological characteristics (endotypes), in order to offer more personalized care. Assigning a patient to any of these groups requires the identification of both clinical and biological markers. Ideally, biological markers should be easily obtained from blood or urine, but these may lack specificity. Biomarkers can be identified initially using conventional or more sophisticated techniques. However, the more sophisticated techniques should be simplified in the future if they are to have clinical utility. The -omics approach offers a methodology that can assist in the investigation and identification of useful markers in both targeted and blind searches. Specifically, metabolomics is the science that studies biological processes involving metabolites, which can be intermediate or final products. The metabolites associated with COPD and their specific phenotypic and endotypic features have been studied using various techniques. Several compounds of particular interest have emerged, namely, several types of lipids and derivatives (mainly phospholipids, but also ceramides, fatty acids and eicosanoids), amino acids, coagulation factors, and nucleic acid components, likely to be involved in their function, protein catabolism, energy production, oxidative stress, immune-inflammatory response and coagulation disorders. However, clear metabolomic profiles of the disease and its various manifestations that may already be applicable in clinical practice still need to be defined.
Collapse
Affiliation(s)
- Joaquim Gea
- Servicio de Neumología, Hospital del Mar - IMIM, Barcelona, Spain; Dpt. MELIS, Universitat Pompeu Fabra, Barcelona, Spain; CIBERES, ISCIII, Barcelona, Spain.
| | - César J Enríquez-Rodríguez
- Servicio de Neumología, Hospital del Mar - IMIM, Barcelona, Spain; Dpt. MELIS, Universitat Pompeu Fabra, Barcelona, Spain
| | - Sergi Pascual-Guardia
- Servicio de Neumología, Hospital del Mar - IMIM, Barcelona, Spain; Dpt. MELIS, Universitat Pompeu Fabra, Barcelona, Spain; CIBERES, ISCIII, Barcelona, Spain
| |
Collapse
|
9
|
Metabolism-Related Gene TXNRD1 Regulates Inflammation and Oxidative Stress Induced by Cigarette Smoke through the Nrf2/HO-1 Pathway in the Small Airway Epithelium. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7067623. [PMID: 36578523 PMCID: PMC9792251 DOI: 10.1155/2022/7067623] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 12/24/2022]
Abstract
Chronic obstructive pulmonary disease (COPD), a small airway disease, is regarded as a metabolic disorder. To further uncover the metabolic profile of COPD patients, it is necessary to identify metabolism-related differential genes in small airway epithelium (SAE) of COPD. Metabolism-related differential genes in SAE between COPD patients and nonsmokers were screened from GSE128708 and GSE20257 datasets. KEGG, GO, and PPI analyses were performed to evaluate the pathway enrichment, term enrichment, and protein interaction of candidate metabolism-related differential genes, respectively. RT-PCR was used to verify the mRNA expression of the top ten differential genes. Western blotting was used to evaluate the protein expression of TXNRD1. TXNRD1 inhibitor auranofin (AUR) was used to assess the impact of TXNRD1 on oxidative stress and inflammation induced by cigarette smoke extraction (CSE). Twenty-four metabolism-related differential genes were selected. ALDH3A1, AKR1C3, CYP1A1, AKC1C1, CPY1B1, and TXNRD1 in the top ten genes were significantly upregulated after CSE simulation for 24 h in human bronchial epithelial (16HBE) cells. Among them, CYP1A1 and TXNRD1 also have a significant upregulation in primary SAE after simulation of CSE for 24 h. The overexpression of protein TXNRD1 has also been detected in 16HBE cells, primary SAE stimulated with CSE, and mouse lung exposed to cigarette smoke (CS). Additionally, inhibition of TXNRD1 with 0.1 μM AUR alleviated the expression of IL-6 and reactive oxygen species (ROS) induced by CSE by activating the Nrf2/HO-1 pathway in 16HBE cells. This study identified twenty-four metabolism-related differential genes associated with COPD. TXNRD1 might participate in the oxidative stress and inflammation induced by CS by regulating the activation of the Nrf2/HO-1 pathway.
Collapse
|
10
|
Goel K, Schweitzer KS, Serban KA, Bittman R, Petrache I. Pharmacological sphingosine-1 phosphate receptor 1 targeting in cigarette smoke-induced emphysema in mice. Am J Physiol Lung Cell Mol Physiol 2022; 322:L794-L803. [PMID: 35412858 PMCID: PMC9109793 DOI: 10.1152/ajplung.00017.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 11/22/2022] Open
Abstract
Primarily caused by chronic cigarette smoking (CS), emphysema is characterized by loss of alveolar cells comprising lung units involved in gas exchange and inflammation that culminate in airspace enlargement. Dysregulation of sphingolipid metabolism with increases of ceramide relative to sphingosine-1 phosphate (S1P) signaling has been shown to cause lung cell apoptosis and is emerging as a potential therapeutic target in emphysema. We sought to determine the impact of augmenting S1P signaling via S1P receptor 1 (S1P1) in a mouse model of CS-induced emphysema. DBA2 mice were exposed to CS for 4 or 6 mo and treated with pharmacological agonists of S1P1: phosphonated FTY720 (FTY720-1S and 2S analogs; 0.01-1.0 mg/kg) or GSK183303A (10 mg/kg). Pharmacological S1P1 agonists ameliorated CS-induced lung parenchymal apoptosis and airspace enlargement as well as loss of body weight. S1P1 agonists had modest inhibitory effects on CS-induced airspace inflammation and lung functional changes measured by Flexivent, improving lung tissue resistance. S1P1 abundance was reduced in chronic CS-conditions and remained decreased after CS-cessation or treatment with FTY720-1S. These results support an important role for S1P-S1P1 axis in maintaining the structural integrity of alveoli during chronic CS exposure and suggest that increasing both S1P1 signaling and abundance may be beneficial to counteract the effects of chronic CS exposure.
Collapse
Affiliation(s)
- Khushboo Goel
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado
| | - Kelly S Schweitzer
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado
- Department of Medicine, Division of Pulmonary and Critical Care, Indiana University, Indianapolis, Indiana
| | - Karina A Serban
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado
- Department of Medicine, Division of Pulmonary and Critical Care, Indiana University, Indianapolis, Indiana
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College City University of New York, Queens, New York
| | - Irina Petrache
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado
- Department of Medicine, Division of Pulmonary and Critical Care, Indiana University, Indianapolis, Indiana
| |
Collapse
|
11
|
Stankovic IN, Colak D. Prenatal Drugs and Their Effects on the Developing Brain: Insights From Three-Dimensional Human Organoids. Front Neurosci 2022; 16:848648. [PMID: 35401083 PMCID: PMC8990163 DOI: 10.3389/fnins.2022.848648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Decades of research have unequivocally demonstrated that fetal exposure to both recreational and prescription drugs in utero negatively impacts the developing brain. More recently, the application of cutting-edge techniques in neurodevelopmental research has attempted to identify how the fetal brain responds to specific environmental stimuli. Meanwhile, human fetal brain studies still encounter ethical considerations and technical limitations in tissue collection. Human-induced pluripotent stem cell (iPSC)-derived brain organoid technology has emerged as a powerful alternative to examine fetal neurobiology. In fact, human 3D organoid tissues recapitulate cerebral development during the first trimester of pregnancy. In this review, we aim to provide a comprehensive summary of fetal brain metabolic studies related to drug abuse in animal and human models. Additionally, we will discuss the current challenges and prospects of using brain organoids for large-scale metabolomics. Incorporating cutting-edge techniques in human brain organoids may lead to uncovering novel molecular and cellular mechanisms of neurodevelopment, direct novel therapeutic approaches, and raise new exciting questions.
Collapse
Affiliation(s)
- Isidora N. Stankovic
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
- *Correspondence: Isidora N. Stankovic,
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
- Gale & Ira Drukier Institute for Children’s Health, Weill Cornell Medicine, Cornell University, New York, NY, United States
- Dilek Colak,
| |
Collapse
|
12
|
Huang Q, Wu X, Gu Y, Wang T, Zhan Y, Chen J, Zeng Z, Lv Y, Zhao J, Xie J. Detection of the Disorders of Glycerophospholipids and Amino Acids Metabolism in Lung Tissue From Male COPD Patients. Front Mol Biosci 2022; 9:839259. [PMID: 35309511 PMCID: PMC8927538 DOI: 10.3389/fmolb.2022.839259] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 01/19/2022] [Indexed: 11/22/2022] Open
Abstract
Background: At present, few studies have reported the metabolic profiles of lung tissue in patients with COPD. Our study attempted to analyze the lung metabolome in male COPD patients and to screen the overlapping biomarkers of the lung and plasma metabolomes. Methods: We performed untargeted metabolomic analysis of normal lung tissue from two independent sets (the discovery set: 20 male COPD patients and 20 controls and the replication set: 47 male COPD patients and 27 controls) and of plasma samples from 80 male subjects containing 40 COPD patients and 40 controls. Results: We found glycerophospholipids (GPs) and Amino acids were the primary classes of differential metabolites between male COPD patients and controls. The disorders of GPs metabolism and the valine, leucine and isoleucine biosynthesis metabolism pathways were identified in lung discovery set and then also validated in the lung replication set. Combining lung tissue and plasma metabolome, Phytosphingosine and l-tryptophan were two overlapping metabolites biomarkers. Binary logistic regression suggested that phytosphingosine together with l-tryptophan was closely associated with male COPD and showed strong diagnostic power with an AUC of 0.911 (95% CI: 0.8460-0.9765). Conclusion: Our study revealed the metabolic perturbations of lung tissues from male COPD patients. The detected disorders of GPs and amino acids may provide an insight into the pathological mechanism of COPD. Phytosphingosine and l-tryptophan were two novel metabolic biomarkers for differentiating COPD patients and controls.
Collapse
Affiliation(s)
- Qian Huang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojie Wu
- Department of Respiratory and Critical Care Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | - Yiya Gu
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Wang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Zhan
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinkun Chen
- Department of Science, Western University, London, ON, Canada
| | - Zhilin Zeng
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongman Lv
- Health Management Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jungang Xie,
| |
Collapse
|
13
|
Paris D, Palomba L, Tramice A, Motta L, Fuschillo S, Maniscalco M, Motta A. Identification of biomarkers in COPD by metabolomics of exhaled breath condensate and serum/plasma. Minerva Med 2022; 113:424-435. [PMID: 35191295 DOI: 10.23736/s0026-4806.22.07957-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third cause of death worldwide, presenting poor long-term outcomes and chronic disability. COPD is a condition with a wide spectrum of clinical presentations because its pathophysiological determinants relate to tobacco smoke, genetic factors, alteration of several metabolic pathways, and oxidative stress. As a consequence, patients present different phenotypes even with comparable degrees of airflow limitation. Because of the increasing social and economic costs of COPD, a growing attention is currently payed to "omics" techniques for more personalized treatments and patient-tailored rehabilitation programs. In this regard, the systematic investigation of the metabolome (i.e., the whole set of endogenous molecules) in biomatrices, namely metabolomics, has become indispensable for phenotyping respiratory diseases. The metabolomic profiling of biological samples contains the small molecules produced during biological processes and their identification and quantification help in the diagnosis, comprehension of disease outcome and treatment response. Exhaled breath condensate (EBC), plasma and serum are biofluids readily available, with negligible invasiveness, and, therefore, suitable for metabolomics investigations. In this paper, we describe the latest advances on metabolomic profiling of EBC, plasma and serum in COPD patients.
Collapse
Affiliation(s)
- Debora Paris
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Napoli, Italy
| | - Letizia Palomba
- Department of Biomolecular Sciences, University Carlo Bo, Urbino, Italy
| | - Annabella Tramice
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Napoli, Italy
| | - Lorenzo Motta
- Section of Radiology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Salvatore Fuschillo
- Pulmonary Rehabilitation Division of the Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Benevento, Italy
| | - Mauro Maniscalco
- Pulmonary Rehabilitation Division of the Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Benevento, Italy
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Napoli, Italy -
| |
Collapse
|
14
|
Fuschillo S, Paris D, Tramice A, Ambrosino P, Palomba L, Maniscalco M, Motta A. Metabolomic profiling of exhaled breath condensate and plasma/serum in chronic obstructive pulmonary disease. Curr Med Chem 2021; 29:2385-2398. [PMID: 34375174 DOI: 10.2174/0929867328666210810122350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/13/2021] [Accepted: 06/17/2021] [Indexed: 11/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is an increasing cause of global morbidity and mortality, with poor long-term outcomes and chronic disability. COPD is a condition with a wide spectrum of clinical presentations, with different phenotypes being identified even among patients with comparable degrees of airflow limitation. Considering the burden of COPD in terms of social and economic costs, in recent years a growing attention has been given to the need of more personalized approaches and patient-tailored rehabilitation programs. In this regard, the systematic analysis of metabolites in biological matrices, namely metabolomics, may become an essential tool in phenotyping diseases. Through the identification and quantification of the small molecules produced during biological processes, metabolomic profiling of biological samples has thus been proposed as an opportunity to identify novel biomarkers of disease outcome and treatment response. Exhaled breath condensate (EBC) and plasma/serum are fluid pools, which can be easily extracted and analyzed. In this review, we discuss the potential clinical applications of the metabolomic profiling of EBC and plasma/serum in COPD.
Collapse
Affiliation(s)
- Salvatore Fuschillo
- Institute Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Division of the Telese Terme Institute, 82037 Telese Terme (BN), Italy
| | - Debora Paris
- Institute of Biomolecular Chemistry, National Research Council, 80078 Pozzuoli (NA), Italy
| | - Annabella Tramice
- Institute of Biomolecular Chemistry, National Research Council, 80078 Pozzuoli (NA), Italy
| | - Pasquale Ambrosino
- Institute Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Division of the Telese Terme Institute, 82037 Telese Terme (BN), Italy
| | - Letizia Palomba
- Department of Biomolecular Sciences, University "Carlo Bo", 61029 Urbino, Italy
| | - Mauro Maniscalco
- Institute Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Division of the Telese Terme Institute, 82037 Telese Terme (BN), Italy
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, 80078 Pozzuoli (NA), Italy
| |
Collapse
|
15
|
Exacerbation of cardiovascular ageing by diabetes mellitus and its associations with acyl-carnitines. Aging (Albany NY) 2021; 13:14785-14805. [PMID: 34088887 PMCID: PMC8221346 DOI: 10.18632/aging.203144] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/17/2021] [Indexed: 12/19/2022]
Abstract
Objective: To demonstrate differences in cardiovascular structure and function between diabetic and non-diabetic older adults. To investigate associations between acyl-carnitines and cardiovascular function as indexed by imaging measurements. Methods: A community-based cohort of older adults without cardiovascular disease underwent current cardiovascular imaging and metabolomics acyl-carnitines profiling based on current and archived sera obtained fifteen years prior to examination. Results: A total of 933 participants (women 56%, n=521) with a mean age 63±13 years were studied. Old diabetics compared to old non-diabetics had lower myocardial relaxation (0.8±0.2 vs 0.9±0.3, p=0.0039); lower left atrial conduit strain (12±4.3 vs 14±4.1, p=0.045), lower left atrial conduit strain rate (-1.2±0.4 vs -1.3±0.5, p=0.042) and lower ratio of left atrial conduit strain to left atrial booster strain (0.5±0.2 vs 0.7±0.3, p=0.0029). Higher levels of archived short chain acyl-carnitine were associated with present-day impairments in myocardial relaxation (C5:1; OR 1.03, p=0.011), worse left atrial conduit strain function (C5:1; OR 1.03, p=0.037). Increases in hydroxylated acyl-carnitines were associated with worse left atrial conduit strain [(C4-OH; OR 1.05, p=0.0017), (C16:2-OH; OR 1.18, p=0.037)]. Current, archived and changes in long chain acyl-carnitines were associated with cardiovascular functions [(C16; OR 1.02, p=0.002), (C20:3; OR 1.01, p=0.014), (C14:3; OR 1.12, p=0.033), (C18:1; OR 1.01, p=0.018), (C18:2; OR 1.01, p=0.028), (C20:4; OR 1.10, p=0.038)] (all p<0.05). Conclusion: Older diabetic adults had significant impairments in left ventricular myocardial relaxation and left atrial strain, compared to older non-diabetic adults. Short chain and long chain, di-carboxyl and hydroxylated acyl-carnitines were associated with these cardiovascular functional differences.
Collapse
|
16
|
Goel K, Beatman EL, Egersdorf N, Scruggs A, Cao D, Berdyshev EV, Schweitzer KS, Petrache I. Sphingosine 1 Phosphate (S1P) Receptor 1 Is Decreased in Human Lung Microvascular Endothelial Cells of Smokers and Mediates S1P Effect on Autophagy. Cells 2021; 10:cells10051200. [PMID: 34068927 PMCID: PMC8156252 DOI: 10.3390/cells10051200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 01/26/2023] Open
Abstract
Destruction of alveoli by apoptosis induced by cigarette smoke (CS) is a major driver of emphysema pathogenesis. However, when compared to cells isolated from non-smokers, primary human lung microvascular endothelial cells (HLMVECs) isolated from chronic smokers are more resilient when exposed to apoptosis-inducing ceramide. Whether this adaptation restores homeostasis is unknown. To better understand the phenotype of HLMVEC in smokers, we interrogated a major pro-survival pathway supported by sphingosine-1-phosphate (S1P) signaling via S1P receptor 1 (S1P1). Primary HLMVECs from lungs of non-smoker or smoker donors were isolated and studied in culture for up to five passages. S1P1 mRNA and protein abundance were significantly decreased in HLMVECs from smokers compared to non-smokers. S1P1 was also decreased in situ in lungs of mice chronically exposed to CS. Levels of S1P1 expression tended to correlate with those of autophagy markers, and increasing S1P (via S1P lyase knockdown with siRNA) stimulated baseline macroautophagy with lysosomal degradation. In turn, loss of S1P1 (siRNA) inhibited these effects of S1P on HLMVECs autophagy. These findings suggest that the anti-apoptotic phenotype of HLMVECs from smokers may be maladaptive, since it is associated with decreased S1P1 expression that may impair their autophagic response to S1P.
Collapse
Affiliation(s)
- Khushboo Goel
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA;
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - Erica L. Beatman
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - Nicholas Egersdorf
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - April Scruggs
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - Danting Cao
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - Evgeny V. Berdyshev
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - Kelly S. Schweitzer
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Irina Petrache
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +1-303-398-1355
| |
Collapse
|
17
|
Koh AS, Kovalik JP. Metabolomics and cardiovascular imaging: a combined approach for cardiovascular ageing. ESC Heart Fail 2021; 8:1738-1750. [PMID: 33783981 PMCID: PMC8120371 DOI: 10.1002/ehf2.13274] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/14/2021] [Accepted: 02/11/2021] [Indexed: 12/18/2022] Open
Abstract
The purpose of this review is to explore how metabolomics can help uncover new biomarkers and mechanisms for cardiovascular ageing. Cardiovascular ageing refers to cardiovascular structural and functional alterations that occur with chronological ageing and that can lead to the development of cardiovascular disease. These alterations, which were previously only detectable on tissue histology or corroborated on blood samples, are now detectable with modern imaging techniques. Despite the emergence of powerful new imaging tools, clinical investigation into cardiovascular ageing is challenging because ageing is a life course phenomenon involving known and unknown risk factors that play out in a dynamic fashion. Metabolomic profiling measures large numbers of metabolites with diverse chemical properties. Metabolomics has the potential to capture changes in biochemistry brought about by pathophysiologic processes as well as by normal ageing. When combined with non-invasive cardiovascular imaging tools, metabolomics can be used to understand pathological consequences of cardiovascular ageing. This review will summarize previous metabolomics and imaging studies in cardiovascular ageing. These methods may be a clinically relevant and novel approach to identify mechanisms of cardiovascular ageing and formulate or personalize treatment strategies.
Collapse
Affiliation(s)
- Angela S Koh
- National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Jean-Paul Kovalik
- Duke-NUS Medical School, Singapore, Singapore.,Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
18
|
Jang YO, Kim OH, Kim SJ, Lee SH, Yun S, Lim SE, Yoo HJ, Shin Y, Lee SW. High-fiber diets attenuate emphysema development via modulation of gut microbiota and metabolism. Sci Rep 2021; 11:7008. [PMID: 33772084 PMCID: PMC7997879 DOI: 10.1038/s41598-021-86404-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
Dietary fiber functions as a prebiotic to determine the gut microbe composition. The gut microbiota influences the metabolic functions and immune responses in human health. The gut microbiota and metabolites produced by various dietary components not only modulate immunity but also impact various organs. Although recent findings have suggested that microbial dysbiosis is associated with several respiratory diseases, including asthma, cystic fibrosis, and allergy, the role of microbiota and metabolites produced by dietary nutrients with respect to pulmonary disease remains unclear. Therefore, we explored whether the gut microbiota and metabolites produced by dietary fiber components could influence a cigarette smoking (CS)-exposed emphysema model. In this study, it was demonstrated that a high-fiber diet including non-fermentable cellulose and fermentable pectin attenuated the pathological changes associated with emphysema progression and the inflammatory response in CS-exposed emphysema mice. Moreover, we observed that different types of dietary fiber could modulate the diversity of gut microbiota and differentially impacted anabolism including the generation of short-chain fatty acids, bile acids, and sphingolipids. Overall, the results of this study indicate that high-fiber diets play a beneficial role in the gut microbiota-metabolite modulation and substantially affect CS-exposed emphysema mice. Furthermore, this study suggests the therapeutic potential of gut microbiota and metabolites from a high-fiber diet in emphysema via local and systemic inflammation inhibition, which may be useful in the development of a new COPD treatment plan.
Collapse
Affiliation(s)
- Yoon Ok Jang
- Department of Pulmonary and Critical Care Medicine, and Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ock-Hwa Kim
- Department of Pulmonary and Critical Care Medicine, and Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Su Jung Kim
- Department of Convergence Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Se Hee Lee
- Department of Pulmonary and Critical Care Medicine, and Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.,Department of Pulmonology, Allergy and Critical Care Medicine, CHA Bundang Medical Center, CHA University, Seongnam-si, Republic of Korea
| | - Sunmi Yun
- Metagenome Service Department, Macrogen, Inc., Seoul, Republic of Korea
| | - Se Eun Lim
- Metagenome Service Department, Macrogen, Inc., Seoul, Republic of Korea
| | - Hyun Ju Yoo
- Department of Convergence Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong Shin
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, and Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
19
|
Aguilar MA, McGuigan J. Semi-automated NMR Pipeline for Environmental Exposures: New Insights on the Metabolomics of Smokers versus Non-smokers. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2021; 26:316-327. [PMID: 33691028 PMCID: PMC8900656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Environmental exposure pathophysiology related to smoking can yield metabolic changes that are difficult to describe in a biologically informative fashion with manual proprietary software. Nuclear magnetic resonance (NMR) spectroscopy detects compounds found in biofluids yielding a metabolic snapshot. We applied our semi-automated NMR pipeline for a secondary analysis of a smoking study (MTBLS374 from the MetaboLights repository) (n = 112). This involved quality control (in the form of data preprocessing), automated metabolite quantification, and analysis. With our approach we putatively identified 79 metabolites that were previously unreported in the dataset. Quantified metabolites were used for metabolic pathway enrichment analysis that replicated 1 enriched pathway with the original study as well as 3 previously unreported pathways. Our pipeline generated a new random forest (RF) classifier between smoking classes that revealed several combinations of compounds. This study broadens our metabolomic understanding of smoking exposure by 1) notably increasing the number of quantified metabolites with our analytic pipeline, 2) suggesting smoking exposure may lead to heterogenous metabolic responses according to random forest modeling, and 3) modeling how newly quantified individual metabolites can determine smoking status. Our approach can be applied to other NMR studies to characterize environmental risk factors, allowing for the discovery of new biomarkers of disease and exposure status.
Collapse
|
20
|
Kasiotis KM, Evergetis E, Papachristos D, Vangelatou O, Antonatos S, Milonas P, Haroutounian SA, Machera K. An essay on ecosystem availability of Nicotiana glauca graham alkaloids: the honeybees case study. BMC Ecol 2020; 20:57. [PMID: 33158433 PMCID: PMC7646078 DOI: 10.1186/s12898-020-00325-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/26/2020] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Invasive plant species pose a significant threat for fragile isolated ecosystems, occupying space, and consuming scarce local resources. Recently though, an additional adverse effect was recognized in the form of its secondary metabolites entering the food chain. The present study is elaborating on this subject with a specific focus on the Nicotiana glauca Graham (Solanaceae) alkaloids and their occurrence and food chain penetrability in Mediterranean ecosystems. For this purpose, a targeted liquid chromatography electrospray tandem mass spectrometric (LC-ESI-MS/MS) analytical method, encompassing six alkaloids and one coumarin derivative, utilizing hydrophilic interaction chromatography (HILIC) was developed and validated. RESULTS The method exhibited satisfactory recoveries, for all analytes, ranging from 75 to 93%, and acceptable repeatability and reproducibility. Four compounds (anabasine, anatabine, nornicotine, and scopoletin) were identified and quantified in 3 N. glauca flowers extracts, establishing them as potential sources of alien bio-molecules. The most abundant constituent was anabasine, determined at 3900 μg/g in the methanolic extract. These extracts were utilized as feeding treatments on Apis mellifera honeybees, resulting in mild toxicity documented by 16-18% mortality. A slightly increased effect was elicited by the methanolic extract containing anabasine at 20 μg/mL, where mortality approached 25%. Dead bees were screened for residues of the N. glauca flower extracts compounds and a significant mean concentration of anabasine was evidenced in both 10 and 20 μg/mL treatments, ranging from 51 to 92 ng/g per bee body weight. Scopoletin was also detected in trace amounts. CONCLUSIONS The mild toxicity of the extracts in conjunction with the alkaloid and coumarin residual detection in bees, suggest that these alien bio-molecules are transferred within the food chain, suggesting a chemical invasion phenomenon, never reported before.
Collapse
Affiliation(s)
- Konstantinos M Kasiotis
- Laboratory of Pesticides' Toxicology, Department of Pesticides Control and Phytopharmacy, Benaki Phytopathological Institute, 8 St. Delta str., 14561, Kifissia, Attica, Greece.
| | - Epameinondas Evergetis
- Laboratory of Nutritional Physiology and Feeding, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece.
| | - Dimitrios Papachristos
- Laboratory of Agricultural Entomology, Department of Entomology and Agricultural Zoology, Benaki Phytopathological Institute, 8 St. Delta str., 14561, Kifissia, Attica, Greece
| | - Olympia Vangelatou
- Laboratory of Nutritional Physiology and Feeding, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece
| | - Spyridon Antonatos
- Laboratory of Agricultural Entomology, Department of Entomology and Agricultural Zoology, Benaki Phytopathological Institute, 8 St. Delta str., 14561, Kifissia, Attica, Greece
| | - Panagiotis Milonas
- Biological Control Laboratory, Department of Entomology and Agricultural Zoology, Benaki Phytopathological Institute, 8 St. Delta str., 14561, Kifissia, Attica, Greece
| | - Serkos A Haroutounian
- Laboratory of Nutritional Physiology and Feeding, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece
| | - Kyriaki Machera
- Laboratory of Pesticides' Toxicology, Department of Pesticides Control and Phytopharmacy, Benaki Phytopathological Institute, 8 St. Delta str., 14561, Kifissia, Attica, Greece
| |
Collapse
|
21
|
Pal A, Al‐Shaer AE, Guesdon W, Torres MJ, Armstrong M, Quinn K, Davis T, Reisdorph N, Neufer PD, Spangenburg EE, Carroll I, Bazinet RP, Halade GV, Clària J, Shaikh SR. Resolvin E1 derived from eicosapentaenoic acid prevents hyperinsulinemia and hyperglycemia in a host genetic manner. FASEB J 2020; 34:10640-10656. [PMID: 32579292 PMCID: PMC7497168 DOI: 10.1096/fj.202000830r] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Abstract
Eicosapentaenoic acid (EPA) has garnered attention after the success of the REDUCE-IT trial, which contradicted previous conclusions on EPA for cardiovascular disease risk. Here we first investigated EPA's preventative role on hyperglycemia and hyperinsulinemia. EPA ethyl esters prevented obesity-induced glucose intolerance, hyperinsulinemia, and hyperglycemia in C57BL/6J mice. Supporting NHANES analyses showed that fasting glucose levels of obese adults were inversely related to EPA intake. We next investigated how EPA improved murine hyperinsulinemia and hyperglycemia. EPA overturned the obesity-driven decrement in the concentration of 18-hydroxyeicosapentaenoic acid (18-HEPE) in white adipose tissue and liver. Treatment of obese inbred mice with RvE1, the downstream immunoresolvant metabolite of 18-HEPE, but not 18-HEPE itself, reversed hyperinsulinemia and hyperglycemia through the G-protein coupled receptor ERV1/ChemR23. To translate the findings, we determined if the effects of RvE1 were dependent on host genetics. RvE1's effects on hyperinsulinemia and hyperglycemia were divergent in diversity outbred mice that model human genetic variation. Secondary SNP analyses further confirmed extensive genetic variation in human RvE1/EPA-metabolizing genes. Collectively, the data suggest EPA prevents hyperinsulinemia and hyperglycemia, in part, through RvE1's activation of ERV1/ChemR23 in a host genetic manner. The studies underscore the need for personalized administration of RvE1 based on genetic/metabolic enzyme profiles.
Collapse
Affiliation(s)
- Anandita Pal
- Department of NutritionGillings School of Global Public Health and School of MedicineThe University of North Carolina at Chapel HillChapel HillNCUSA
| | - Abrar E. Al‐Shaer
- Department of NutritionGillings School of Global Public Health and School of MedicineThe University of North Carolina at Chapel HillChapel HillNCUSA
| | - William Guesdon
- Department of Biochemistry & Molecular BiologyBrody School of MedicineEast Carolina UniversityGreenvilleNCUSA
- Present address:
School of Immunology and Microbial SciencesKing's College LondonGuy's CampusLondonSE1 9RTUK
| | - Maria J. Torres
- Department of PhysiologyEast Carolina Diabetes & Obesity InstituteEast Carolina UniversityGreenvilleNCUSA
- Present address:
Duke Molecular Physiology InstituteDuke University300 North Duke StreetDurhamNC27701USA
| | - Michael Armstrong
- Department of Pharmaceutical SciencesUniversity of Colorado Denver Anschutz Medical CampusAuroraCOUSA
| | - Kevin Quinn
- Department of Pharmaceutical SciencesUniversity of Colorado Denver Anschutz Medical CampusAuroraCOUSA
| | - Traci Davis
- Department of NutritionGillings School of Global Public Health and School of MedicineThe University of North Carolina at Chapel HillChapel HillNCUSA
| | - Nichole Reisdorph
- Department of Pharmaceutical SciencesUniversity of Colorado Denver Anschutz Medical CampusAuroraCOUSA
| | - P. Darrell Neufer
- Department of PhysiologyEast Carolina Diabetes & Obesity InstituteEast Carolina UniversityGreenvilleNCUSA
| | - Espen E. Spangenburg
- Department of PhysiologyEast Carolina Diabetes & Obesity InstituteEast Carolina UniversityGreenvilleNCUSA
| | - Ian Carroll
- Department of NutritionGillings School of Global Public Health and School of MedicineThe University of North Carolina at Chapel HillChapel HillNCUSA
| | | | - Ganesh V. Halade
- Division of Cardiovascular SciencesDepartment of MedicineThe University of South FloridaTampaFLUSA
| | - Joan Clària
- Department of Biochemistry and Molecular GeneticsUniversity of BarcelonaHospital ClínicBarcelonaSpain
| | - Saame Raza Shaikh
- Department of NutritionGillings School of Global Public Health and School of MedicineThe University of North Carolina at Chapel HillChapel HillNCUSA
| |
Collapse
|
22
|
Nutrimetabolomics reveals food-specific compounds in urine of adults consuming a DASH-style diet. Sci Rep 2020; 10:1157. [PMID: 31980691 PMCID: PMC6981146 DOI: 10.1038/s41598-020-57979-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/08/2020] [Indexed: 01/22/2023] Open
Abstract
Although health benefits of the Dietary Approaches to Stop Hypertension (DASH) diet are established, it is not understood which food compounds result in these benefits. We used metabolomics to identify unique compounds from individual foods of a DASH-style diet and determined if these Food-Specific Compounds (FSC) are detectable in urine from participants in a DASH-style dietary study. We also examined relationships between urinary compounds and blood pressure (BP). Nineteen subjects were randomized into 6-week controlled DASH-style diet interventions. Mass spectrometry-based metabolomics was performed on 24-hour urine samples collected before and after each intervention and on 12 representative DASH-style foods. Between 66–969 compounds were catalogued as FSC; for example, 4-hydroxydiphenylamine was found to be unique to apple. Overall, 13–190 of these FSC were detected in urine, demonstrating that these unmetabolized food compounds can be discovered in urine using metabolomics. Although linear mixed effects models showed no FSC from the 12 profiled foods were significantly associated with BP, other endogenous and food-related compounds were associated with BP (N = 16) and changes in BP over time (N = 6). Overall, this proof of principle study demonstrates that metabolomics can be used to catalog FSC, which can be detected in participant urine following a dietary intervention.
Collapse
|
23
|
Reisdorph R, Michel C, Quinn K, Doenges K, Reisdorph N. Untargeted Differential Metabolomics Analysis Using Drift Tube Ion Mobility-Mass Spectrometry. Methods Mol Biol 2020; 2084:55-78. [PMID: 31729653 DOI: 10.1007/978-1-0716-0030-6_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mass spectrometry-based metabolomics is being increasingly applied to a number of applications, including the fields of clinical, industrial, plant, and nutritional science. Several improvements have advanced the field considerably over the past decade, including ultra-high performance liquid chromatography (uHPLC), column chemistries, instruments, software, and molecular databases. However, challenges remain, including how to separate small molecules that are part of highly complex samples; this can be accomplished using chromatographic techniques or through improved resolution in the gas phase. Ion mobility-mass spectrometry (IM-MS) provides an extra dimension of gas phase separation that can result in improvements to both quantitation and compound identification. Here we describe a typical drift tube IM-MS metabolomics workflow, which includes the following steps: (1) Data acquisition, (2) Data preprocessing, (3) Molecular feature finding, and (4) Differential analysis and Molecular annotation. Overall, these methods can help investigators from a variety of scientific fields use IM-MS metabolomics as part of their own workflow.
Collapse
Affiliation(s)
- Rick Reisdorph
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA.
| | - Cole Michel
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Kevin Quinn
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Katrina Doenges
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| |
Collapse
|
24
|
Rizvi A, Shankar A, Chatterjee A, More TH, Bose T, Dutta A, Balakrishnan K, Madugulla L, Rapole S, Mande SS, Banerjee S, Mande SC. Rewiring of Metabolic Network in Mycobacterium tuberculosis During Adaptation to Different Stresses. Front Microbiol 2019; 10:2417. [PMID: 31736886 PMCID: PMC6828651 DOI: 10.3389/fmicb.2019.02417] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/07/2019] [Indexed: 12/15/2022] Open
Abstract
Metabolic adaptation of Mycobacterium tuberculosis (M. tuberculosis) to microbicidal intracellular environment of host macrophages is fundamental to its pathogenicity. However, an in-depth understanding of metabolic adjustments through key reaction pathways and networks is limited. To understand how such changes occur, we measured the cellular metabolome of M. tuberculosis subjected to four microbicidal stresses using liquid chromatography-mass spectrometric multiple reactions monitoring (LC-MRM/MS). Overall, 87 metabolites were identified. The metabolites best describing the separation between stresses were identified through multivariate analysis. The coupling of the metabolite measurements with existing genome-scale metabolic model, and using constraint-based simulation led to several new concepts and unreported observations in M. tuberculosis; such as (i) the high levels of released ammonia as an adaptive response to acidic stress was due to increased flux through L-asparaginase rather than urease activity; (ii) nutrient starvation-induced anaplerotic pathway for generation of TCA intermediates from phosphoenolpyruvate using phosphoenolpyruvate kinase; (iii) quenching of protons through GABA shunt pathway or sugar alcohols as possible mechanisms of early adaptation to acidic and oxidative stresses; and (iv) usage of alternate cofactors by the same enzyme as a possible mechanism of rewiring metabolic pathways to overcome stresses. Besides providing new leads and important nodes that can be used for designing intervention strategies, the study advocates the strength of applying flux balance analyses coupled with metabolomics to get a global picture of complex metabolic adjustments.
Collapse
Affiliation(s)
- Arshad Rizvi
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Arvind Shankar
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| | | | | | - Tungadri Bose
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| | - Anirban Dutta
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| | - Kannan Balakrishnan
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Lavanya Madugulla
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | | | - Sharmila S Mande
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| | - Sharmistha Banerjee
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | | |
Collapse
|
25
|
An Updated Overview of Metabolomic Profile Changes in Chronic Obstructive Pulmonary Disease. Metabolites 2019; 9:metabo9060111. [PMID: 31185592 PMCID: PMC6631716 DOI: 10.3390/metabo9060111] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD), a common and heterogeneous respiratory disease, is characterized by persistent and incompletely reversible airflow limitation. Metabolomics is applied to analyze the difference of metabolic profile based on the low-molecular-weight metabolites (<1 kDa). Emerging metabolomic analysis may provide insights into the pathogenesis and diagnosis of COPD. This review aims to summarize the alteration of metabolites in blood/serum/plasma, urine, exhaled breath condensate, lung tissue samples, etc. from COPD individuals, thereby uncovering the potential pathogenesis of COPD according to the perturbed metabolic pathways. Metabolomic researches have indicated that the dysfunctions of amino acid metabolism, lipid metabolism, energy production pathways, and the imbalance of oxidations and antioxidations might lead to local and systematic inflammation by activating the Nuclear factor kappa-light-chain-enhancer of activated B cells signaling pathway and releasing inflammatory cytokines, like interleutin-6 (IL-6), tumor necrosis factor-α, and IL-8. In addition, they might cause protein malnutrition and oxidative stress and contribute to the development and exacerbation of COPD.
Collapse
|
26
|
Mu L, Niu Z, Blair RH, Yu H, Browne RW, Bonner MR, Fanter T, Deng F, Swanson M. Metabolomics Profiling before, during, and after the Beijing Olympics: A Panel Study of Within-Individual Differences during Periods of High and Low Air Pollution. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:57010. [PMID: 31140880 PMCID: PMC6791568 DOI: 10.1289/ehp3705] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 04/03/2019] [Accepted: 04/22/2019] [Indexed: 05/19/2023]
Abstract
BACKGROUND The metabolome is a collection of exogenous chemicals and metabolites from cellular processes that may reflect the body's response to environmental exposures. Studies of air pollution and metabolomics are limited. OBJECTIVES To explore changes in the human metabolome before, during, and after the 2008 Beijing Olympics Games, when air pollution was high, low, and high, respectively. METHODS Serum samples were collected before, during, and after the Olympics from 26 participants in an existing panel study. Gas and ultra-high performance liquid chromatography/mass spectrometry were used in metabolomics analysis. Repeated measures ANOVA, network analysis, and enrichment analysis methods were employed to identify metabolites and classes associated with air pollution changes. RESULTS A total of 886 molecules were measured in our metabolomics analysis. Network partitioning identified four modules with 65 known metabolites that significantly changed across the three time points. All known molecules in the first module ([Formula: see text]) were lipids (e.g., eicosapentaenoic acid, stearic acid). The second module consisted primarily of dipeptides ([Formula: see text], e.g., isoleucylglycine) plus 8 metabolites from four other classes (e.g., hypoxanthine, 12-hydroxyeicosatetraenoic acid). Most of the metabolites in Modules 3 (19 of 23) and 4 (5 of 5) were unknown. Enrichment analysis of module-identified metabolites indicted significantly overrepresented pathways, including long- and medium-chain fatty acids, polyunsaturated fatty acids (n3 and n6), eicosanoids, lysolipid, dipeptides, fatty acid metabolism, and purine metabolism [(hypo) xanthine/inosine-containing pathways]. CONCLUSIONS We identified two major metabolic signatures: one consisting of lipids, and a second that included dipeptides, polyunsaturated fatty acids, taurine, and xanthine. Metabolites in both groups decreased during the 2008 Beijing Olympics, when air pollution was low, and increased after the Olympics, when air pollution returned to normal (high) levels. https://doi.org/10.1289/EHP3705.
Collapse
Affiliation(s)
- Lina Mu
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Zhongzheng Niu
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Rachael Hageman Blair
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Han Yu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Richard W. Browne
- Department of Biotechnical and Clinical Laboratory Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Matthew R. Bonner
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Tiffany Fanter
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Furong Deng
- Department of Occupational and Environmental Health, School of Public Health, Peking University, Beijing, China
| | - Mya Swanson
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, New York, USA
| |
Collapse
|
27
|
An Official American Thoracic Society Workshop Report: Obesity and Metabolism. An Emerging Frontier in Lung Health and Disease. Ann Am Thorac Soc 2018; 14:1050-1059. [PMID: 28570148 DOI: 10.1513/annalsats.201703-263ws] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The world is in the midst of an unprecedented epidemic of obesity. This epidemic has changed the presentation and etiology of common diseases. For example, steatohepatitis, directly attributable to obesity, is now the most common cause of cirrhosis in the United States. Type 2 diabetes is increasingly being diagnosed in children. Pulmonary researchers and clinicians are just beginning to appreciate the impact of obesity and altered metabolism on common pulmonary diseases. Obesity has recently been identified as a major risk factor for the development of asthma and for acute respiratory distress syndrome. Obesity is associated with profound changes in pulmonary physiology, the development of pulmonary hypertension, sleep-disordered breathing, and altered susceptibility to pulmonary infection. In short, obesity is leading to dramatic changes in lung health and disease. Simultaneously, the rapidly developing field of metabolism, including mitochondrial function, is shifting the paradigms by which the pathophysiology of many pulmonary diseases is understood. Altered metabolism can lead to profound changes in both innate and adaptive immunity, as well as the function of structural cells. To address this emerging field, a 3-day meeting on obesity, metabolism, and lung disease was convened in October 2015 to discuss recent findings, foster research initiatives, and ultimately guide clinical care. The major findings arising from this meeting are reported in this document.
Collapse
|
28
|
Walmsley S, Cruickshank-Quinn C, Quinn K, Zhang X, Petrache I, Bowler RP, Reisdorph R, Reisdorph N. A prototypic small molecule database for bronchoalveolar lavage-based metabolomics. Sci Data 2018; 5:180060. [PMID: 29664467 PMCID: PMC5903367 DOI: 10.1038/sdata.2018.60] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 02/09/2018] [Indexed: 02/06/2023] Open
Abstract
The analysis of bronchoalveolar lavage fluid (BALF) using mass spectrometry-based metabolomics can provide insight into lung diseases, such as asthma. However, the important step of compound identification is hindered by the lack of a small molecule database that is specific for BALF. Here we describe prototypic, small molecule databases derived from human BALF samples (n=117). Human BALF was extracted into lipid and aqueous fractions and analyzed using liquid chromatography mass spectrometry. Following filtering to reduce contaminants and artifacts, the resulting BALF databases (BALF-DBs) contain 11,736 lipid and 658 aqueous compounds. Over 10% of these were found in 100% of samples. Testing the BALF-DBs using nested test sets produced a 99% match rate for lipids and 47% match rate for aqueous molecules. Searching an independent dataset resulted in 45% matching to the lipid BALF-DB compared to<25% when general databases are searched. The BALF-DBs are available for download from MetaboLights. Overall, the BALF-DBs can reduce false positives and improve confidence in compound identification compared to when general databases are used.
Collapse
Affiliation(s)
- Scott Walmsley
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Charmion Cruickshank-Quinn
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kevin Quinn
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Xing Zhang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Irina Petrache
- Department of Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - Russell P Bowler
- Department of Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - Richard Reisdorph
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nichole Reisdorph
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
29
|
Reisdorph NA, Cruickshank-Quinn C, Nkrumah-Elie Y, Reisdorph R. Application of Metabolomics in Lung Research. Methods Mol Biol 2018; 1809:263-288. [PMID: 29987794 DOI: 10.1007/978-1-4939-8570-8_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Advancements in omics technologies have increased our potential to evaluate molecular changes in a rapid and comprehensive manner. This is especially true in mass spectrometry-based metabolomics where improvements, including ease of use, in high-performance liquid chromatography (HPLC), column chemistries, instruments, software, and molecular databases, have advanced the field considerably. Applications of this relatively new omics technology in clinical research include discovering disease biomarkers, finding new drug targets, and elucidating disease mechanisms. Here we describe a typical clinical metabolomics workflow, which includes the following steps: (1) extraction of metabolites from the lung, plasma, bronchoalveolar lavage, or cells; (2) sample analysis via liquid chromatography-mass spectrometry; and (3) data analysis using commercial and freely available software packages. Overall, the methods delineated here can help investigators use metabolomics to discovery novel biomarkers and to understand lung diseases.
Collapse
Affiliation(s)
- Nichole A Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | | | - Yasmeen Nkrumah-Elie
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Richard Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
30
|
Quinn KD, Schedel M, Nkrumah-Elie Y, Joetham A, Armstrong M, Cruickshank-Quinn C, Reisdorph R, Gelfand EW, Reisdorph N. Dysregulation of metabolic pathways in a mouse model of allergic asthma. Allergy 2017; 72:1327-1337. [PMID: 28213886 DOI: 10.1111/all.13144] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Asthma is a complex lung disease resulting from the interplay of genetic and environmental factors. To understand the molecular changes that occur during the development of allergic asthma without genetic and environmental confounders, an experimental model of allergic asthma in mice was used. Our goals were to (1) identify changes at the small molecule level due to allergen exposure, (2) determine perturbed pathways due to disease, and (3) determine whether small molecule changes correlate with lung function. METHODS In this experimental model of allergic asthma, matched bronchoalveolar lavage (BAL) fluid and plasma were collected from three groups of C57BL6 mice (control vs sensitized and/or challenged with ovalbumin, n=3-5/group) 6 hour, 24 hour, and 48 hour after the last challenge. Samples were analyzed using liquid chromatography-mass spectrometry-based metabolomics. Airway hyper-responsiveness (AHR) measurements and differential cell counts were performed. RESULTS In total, 398 and 368 dysregulated metabolites in the BAL fluid and plasma of sensitized and challenged mice were identified, respectively. These belonged to four, interconnected pathways relevant to asthma pathogenesis: sphingolipid metabolism (P=6.6×10-5 ), arginine and proline metabolism (P=1.12×10-7 ), glycerophospholipid metabolism (P=1.3×10-10 ), and the neurotrophin signaling pathway (P=7.0×10-6 ). Furthermore, within the arginine and proline metabolism pathway, a positive correlation between urea-1-carboxylate and AHR was observed in plasma metabolites, while ornithine revealed a reciprocal effect. In addition, agmatine positively correlated with lung eosinophilia. CONCLUSION These findings point to potential targets and pathways that may be central to asthma pathogenesis and can serve as novel therapeutic targets.
Collapse
Affiliation(s)
- K. D. Quinn
- School of Pharmacy and Pharmaceutical Sciences; University of Colorado Denver; Aurora CO USA
- Immunology & Microbiology Department School of Medicine; University of Colorado Denver; Aurora CO USA
| | - M. Schedel
- Division of Cell Biology; Department of Pediatrics; National Jewish Health; Denver CO USA
| | - Y. Nkrumah-Elie
- School of Pharmacy and Pharmaceutical Sciences; University of Colorado Denver; Aurora CO USA
| | - A. Joetham
- Division of Cell Biology; Department of Pediatrics; National Jewish Health; Denver CO USA
| | - M. Armstrong
- School of Pharmacy and Pharmaceutical Sciences; University of Colorado Denver; Aurora CO USA
| | - C. Cruickshank-Quinn
- School of Pharmacy and Pharmaceutical Sciences; University of Colorado Denver; Aurora CO USA
| | - R. Reisdorph
- School of Pharmacy and Pharmaceutical Sciences; University of Colorado Denver; Aurora CO USA
| | - E. W. Gelfand
- Division of Cell Biology; Department of Pediatrics; National Jewish Health; Denver CO USA
| | - N. Reisdorph
- School of Pharmacy and Pharmaceutical Sciences; University of Colorado Denver; Aurora CO USA
- Immunology & Microbiology Department School of Medicine; University of Colorado Denver; Aurora CO USA
| |
Collapse
|
31
|
Metabolomic similarities between bronchoalveolar lavage fluid and plasma in humans and mice. Sci Rep 2017; 7:5108. [PMID: 28698669 PMCID: PMC5505974 DOI: 10.1038/s41598-017-05374-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
This observational study catalogues the overlap in metabolites between matched bronchoalveolar lavage fluid (BALF) and plasma, identifies the degree of congruence between these metabolomes in human and mouse, and determines how molecules may change in response to cigarette smoke (CS) exposure. Matched BALF and plasma was collected from mice (ambient air or CS-exposed) and humans (current or former smokers), and analyzed using mass spectrometry. There were 1155 compounds in common in all 4 sample types; fatty acyls and glycerophospholipids strongly overlapped between groups. In humans and mice, more than half of the metabolites present in BALF were also present in plasma. Mouse BALF and human BALF had a strong positive correlation with 2040 metabolites in common, suggesting that mouse models can be used to interrogate human lung metabolome changes. While power was affected by small sample size in the mouse study, the BALF metabolome appeared to be more affected by CS than plasma. CS-exposed mice showed increased plasma and BALF glycerolipids and glycerophospholipids. This is the first report cataloguing the metabolites present across mouse and human, BALF and plasma. Findings are relevant to translational studies where mouse models are used to examine human disease, and where plasma may be interrogated in lieu of BALF or lung tissue.
Collapse
|
32
|
Gene and metabolite time-course response to cigarette smoking in mouse lung and plasma. PLoS One 2017; 12:e0178281. [PMID: 28575117 PMCID: PMC5456044 DOI: 10.1371/journal.pone.0178281] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 05/10/2017] [Indexed: 12/15/2022] Open
Abstract
Prolonged cigarette smoking (CS) causes chronic obstructive pulmonary disease (COPD), a prevalent serious condition that may persist or progress after smoking cessation. To provide insight into how CS triggers COPD, we investigated temporal patterns of lung transcriptome expression and systemic metabolome changes induced by chronic CS exposure and smoking cessation. Whole lung RNA-seq data was analyzed at transcript and exon levels from C57Bl/6 mice exposed to CS for 1- or 7 days, for 3-, 6-, or 9 months, or for 6 months followed by 3 months of cessation using age-matched littermate controls. We identified previously unreported dysregulation of pyrimidine metabolism and phosphatidylinositol signaling pathways and confirmed alterations in glutathione metabolism and circadian gene pathways. Almost all dysregulated pathways demonstrated reversibility upon smoking cessation, except the lysosome pathway. Chronic CS exposure was significantly linked with alterations in pathways encoding for energy, phagocytosis, and DNA repair and triggered differential expression of genes or exons previously unreported to associate with CS or COPD, including Lox, involved in matrix remodeling, Gp2, linked to goblet cells, and Slc22a12 and Agpat3, involved in purine and glycerolipid metabolism, respectively. CS-induced lung metabolic pathways changes were validated using metabolomic profiles of matched plasma samples, indicating that dynamic metabolic gene regulation caused by CS is reflected in the plasma metabolome. Using advanced technologies, our study uncovered novel pathways and genes altered by chronic CS exposure, including those involved in pyrimidine metabolism, phosphatidylinositol signaling and lysosome function, highlighting their potential importance in the pathogenesis or diagnosis of CS-associated conditions.
Collapse
|
33
|
Justice JN, Johnson LC, DeVan AE, Cruickshank-Quinn C, Reisdorph N, Bassett CJ, Evans TD, Brooks FA, Bryan NS, Chonchol MB, Giordano T, McQueen MB, Seals DR. Improved motor and cognitive performance with sodium nitrite supplementation is related to small metabolite signatures: a pilot trial in middle-aged and older adults. Aging (Albany NY) 2016; 7:1004-21. [PMID: 26626856 PMCID: PMC4694069 DOI: 10.18632/aging.100842] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Advancing age is associated with reductions in nitric oxide bioavailability and changes in metabolic activity, which are implicated in declines in motor and cognitive function. In preclinical models, sodium nitrite supplementation (SN) increases plasma nitrite and improves motor function, whereas other nitric oxide-boosting agents improve cognitive function. This pilot study was designed to translate these findings to middle-aged and older (MA/O) humans to provide proof-of-concept support for larger trials. SN (10 weeks, 80 or 160 mg/day capsules, TheraVasc, Inc.) acutely and chronically increased plasma nitrite and improved performance on measures of motor and cognitive outcomes (all p<0.05 or better) in healthy MA/O adults (62 ± 7 years). Untargeted metabolomics analysis revealed that SN significantly altered 33 (160 mg/day) to 45 (80 mg/day) different metabolites, 13 of which were related to changes in functional outcomes; baseline concentrations of 99 different metabolites predicted functional improvements with SN. This pilot study provides the first evidence that SN improves aspects of motor and cognitive function in healthy MA/O adults, and that these improvements are associated with, and predicted by, the plasma metabolome. Our findings provide the necessary support for larger clinical trials on this promising pharmacological strategy for preserving physiological function with aging.
Collapse
Affiliation(s)
- Jamie N Justice
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Lawrence C Johnson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Allison E DeVan
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Charmion Cruickshank-Quinn
- Integrated Department of Immunology, University of Colorado Anschutz Medical Campus and National Jewish Hospital, Denver, CO 80045, USA
| | - Nichole Reisdorph
- Integrated Department of Immunology, University of Colorado Anschutz Medical Campus and National Jewish Hospital, Denver, CO 80045, USA
| | - Candace J Bassett
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Trent D Evans
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Forrest A Brooks
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | | | - Michel B Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO 80045, USA
| | | | - Matthew B McQueen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
34
|
Heischmann S, Quinn K, Cruickshank-Quinn C, Liang LP, Reisdorph R, Reisdorph N, Patel M. Exploratory Metabolomics Profiling in the Kainic Acid Rat Model Reveals Depletion of 25-Hydroxyvitamin D3 during Epileptogenesis. Sci Rep 2016; 6:31424. [PMID: 27526857 PMCID: PMC4985632 DOI: 10.1038/srep31424] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/20/2016] [Indexed: 12/02/2022] Open
Abstract
Currently, no reliable markers are available to evaluate the epileptogenic potential of a brain injury. The electroencephalogram is the standard method of diagnosis of epilepsy; however, it is not used to predict the risk of developing epilepsy. Biomarkers that indicate an individual's risk to develop epilepsy, especially those measurable in the periphery are urgently needed. Temporal lobe epilepsy (TLE), the most common form of acquired epilepsy, is characterized by spontaneous recurrent seizures following brain injury and a seizure-free "latent" period. Elucidation of mechanisms at play during epilepsy development (epileptogenesis) in animal models of TLE could enable the identification of predictive biomarkers. Our pilot study using liquid chromatography-mass spectrometry metabolomics analysis revealed changes (p-value ≤ 0.05, ≥1.5-fold change) in lipid, purine, and sterol metabolism in rat plasma and hippocampus during epileptogenesis and chronic epilepsy in the kainic acid model of TLE. Notably, disease development was associated with dysregulation of vitamin D3 metabolism at all stages and plasma 25-hydroxyvitamin D3 depletion in the acute and latent phase of injury-induced epileptogenesis. These data suggest that plasma VD3 metabolites reflect the severity of an epileptogenic insult and that a panel of plasma VD3 metabolites may be able to serve as a marker of epileptogenesis.
Collapse
Affiliation(s)
- Svenja Heischmann
- Department of Pharmaceutical Sciences, University of Colorado, School of Pharmacy, 12850 East Montview Boulevard, Aurora, CO 80045, USA
- Department of Immunology, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Kevin Quinn
- Department of Immunology, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | | | - Li-Ping Liang
- Department of Pharmaceutical Sciences, University of Colorado, School of Pharmacy, 12850 East Montview Boulevard, Aurora, CO 80045, USA
| | - Rick Reisdorph
- Department of Immunology, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Nichole Reisdorph
- Department of Immunology, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado, School of Pharmacy, 12850 East Montview Boulevard, Aurora, CO 80045, USA
| |
Collapse
|
35
|
Metabolomics screening identifies reduced L-carnitine to be associated with progressive emphysema. Clin Sci (Lond) 2016; 130:273-87. [DOI: 10.1042/cs20150438] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/12/2015] [Indexed: 01/29/2023]
Abstract
The progression of emphysema, a severe chronic lung disease, was found to be associated with reduced lung tissue-specific L-carnitine in a clinically relevant mouse model. Furthermore, supplementing mice with this metabolite improved lung function and impaired disease progression.
Collapse
|
36
|
Petrache I, Berdyshev EV. Ceramide Signaling and Metabolism in Pathophysiological States of the Lung. Annu Rev Physiol 2015; 78:463-80. [PMID: 26667073 DOI: 10.1146/annurev-physiol-021115-105221] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Following the discovery of ceramide as the central signaling and metabolic relay among sphingolipids, studies of its involvement in lung health and pathophysiology have exponentially increased. In this review, we highlight key studies in the context of recent progress in metabolomics and translational research methodologies. Evidence points toward an important role for the ceramide/sphingosine-1-phosphate rheostat in maintaining lung cell survival, vascular barrier function, and proper host response to airway microbial infections. Sphingosine kinase 1 has emerged as an important determinant of sphingosine-1-phosphate lung levels, which, when aberrantly high, contribute to lung fibrosis, maladaptive vascular remodeling, and allergic asthma. New sphingolipid metabolites have been discovered as potential biomarkers of several lung diseases. Although multiple acute and chronic lung pathological conditions involve perturbations in sphingolipid signaling and metabolism, there are specific patterns, unique sphingolipid species, enzymes, metabolites, and receptors, which have emerged that deepen our understanding of lung pathophysiology and inform the development of new therapies for lung diseases.
Collapse
Affiliation(s)
- Irina Petrache
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, National Jewish Health, Denver, Colorado 80206; ,
| | - Evgeny V Berdyshev
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, National Jewish Health, Denver, Colorado 80206; ,
| |
Collapse
|
37
|
Barrios C, Beaumont M, Pallister T, Villar J, Goodrich JK, Clark A, Pascual J, Ley RE, Spector TD, Bell JT, Menni C. Gut-Microbiota-Metabolite Axis in Early Renal Function Decline. PLoS One 2015; 10:e0134311. [PMID: 26241311 PMCID: PMC4524635 DOI: 10.1371/journal.pone.0134311] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/07/2015] [Indexed: 01/15/2023] Open
Abstract
Introduction Several circulating metabolites derived from bacterial protein fermentation have been found to be inversely associated with renal function but the timing and disease severity is unclear. The aim of this study is to explore the relationship between indoxyl-sulfate, p-cresyl-sulfate, phenylacetylglutamine and gut-microbial profiles in early renal function decline. Results Indoxyl-sulfate (Beta(SE) = -2.74(0.24); P = 8.8x10-29), p-cresyl-sulfate (-1.99(0.24), P = 4.6x10-16), and phenylacetylglutamine(-2.73 (0.25), P = 1.2x10-25) were inversely associated with eGFR in a large population base cohort (TwinsUK, n = 4439) with minimal renal function decline. In a sub-sample of 855 individuals, we analysed metabolite associations with 16S gut microbiome profiles (909 profiles, QIIME 1.7.0). Three Operational Taxonomic Units (OTUs) were significantly associated with indoxyl-sulfate and 52 with phenylacetylglutamine after multiple testing; while one OTU was nominally associated with p-cresyl sulfate. All 56 microbial members belong to the order Clostridiales and are represented by anaerobic Gram-positive families Christensenellaceae, Ruminococcaceae and Lachnospiraceae. Within these, three microbes were also associated with eGFR. Conclusions Our data suggest that indoxyl-sulfate, p-cresyl-sulfate and phenylacetylglutamine are early markers of renal function decline. Changes in the intestinal flora associated with these metabolites are detectable in early kidney disease. Future efforts should dissect this relationship to improve early diagnostics and therapeutics strategies.
Collapse
Affiliation(s)
- Clara Barrios
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
- Department of Nephrology, Hospital del Mar, Institut Mar d’Investigacions Mediques, Barcelona, Spain
- * E-mail: (CB); (CM)
| | - Michelle Beaumont
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Tess Pallister
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Judith Villar
- Department of Infectious Diseases, Hospital del Mar, Institut Mar d’Investigacions Mediques, Barcelona, Spain
| | - Julia K. Goodrich
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States of America
| | - Andrew Clark
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States of America
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar, Institut Mar d’Investigacions Mediques, Barcelona, Spain
| | - Ruth E. Ley
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States of America
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Jordana T. Bell
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
- * E-mail: (CB); (CM)
| |
Collapse
|
38
|
Lockett AD, Petrusca DN, Justice MJ, Poirier C, Serban KA, Rush NI, Kamocka M, Predescu D, Predescu S, Petrache I. Scavenger receptor class B, type I-mediated uptake of A1AT by pulmonary endothelial cells. Am J Physiol Lung Cell Mol Physiol 2015; 309:L425-34. [PMID: 26092999 DOI: 10.1152/ajplung.00376.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 06/09/2015] [Indexed: 12/13/2022] Open
Abstract
In addition to exerting a potent anti-elastase function, α-1 antitrypsin (A1AT) maintains the structural integrity of the lung by inhibiting endothelial inflammation and apoptosis. A main serpin secreted in circulation by hepatocytes, A1AT requires uptake by the endothelium to achieve vasculoprotective effects. This active uptake mechanism, which is inhibited by cigarette smoking (CS), involves primarily clathrin- but also caveola-mediated endocytosis and may require active binding to a receptor. Because circulating A1AT binds to high-density lipoprotein (HDL), we hypothesized that scavenging receptors are candidates for endothelial uptake of the serpin. Although the low-density lipoprotein (LDL) receptor-related protein 1 (LRP1) internalizes only elastase-bound A1AT, the scavenger receptor B type I (SR-BI), which binds and internalizes HDL and is modulated by CS, may be involved in A1AT uptake. Transmission electron microscopy imaging of colloidal gold-labeled A1AT confirmed A1AT endocytosis in both clathrin-coated vesicles and caveolae in endothelial cells. SR-BI immunoprecipitation identified binding to A1AT at the plasma membrane. Pretreatment of human lung microvascular endothelial cells with SR-B ligands (HDL or LDL), knockdown of SCARB1 expression, or neutralizing SR-BI antibodies significantly reduced A1AT uptake by 30-50%. Scarb1 null mice exhibited decreased A1AT lung content following systemic A1AT administration and reduced lung anti-inflammatory effects of A1AT supplementation during short-term CS exposure. In turn, A1AT supplementation increased lung SR-BI expression and modulated circulating lipoprotein levels in wild-type animals. These studies indicate that SR-BI is an important mediator of A1AT endocytosis in pulmonary endothelium and suggest a cross talk between A1AT and lipoprotein regulation of vascular functions.
Collapse
Affiliation(s)
- Angelia D Lockett
- Division of Pulmonary, Allergy, Critical Care and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana;
| | - Daniela N Petrusca
- Division of Pulmonary, Allergy, Critical Care and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Matthew J Justice
- Division of Pulmonary, Allergy, Critical Care and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Christophe Poirier
- Division of Pulmonary, Allergy, Critical Care and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Karina A Serban
- Division of Pulmonary, Allergy, Critical Care and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Natalia I Rush
- Division of Pulmonary, Allergy, Critical Care and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Malgorzata Kamocka
- Division of Nephrology, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Dan Predescu
- Department of Pharmacology, Rush University, Chicago, Illinois; and
| | - Sanda Predescu
- Department of Pharmacology, Rush University, Chicago, Illinois; and
| | - Irina Petrache
- Division of Pulmonary, Allergy, Critical Care and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana; The Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| |
Collapse
|