1
|
Singh A, Siddiqui MA, Pandey S, Azim A, Sinha N. Unveiling Pathophysiological Insights: Serum Metabolic Dysregulation in Acute Respiratory Distress Syndrome Patients with Acute Kidney Injury. J Proteome Res 2024; 23:4216-4228. [PMID: 39078945 DOI: 10.1021/acs.jproteome.4c00138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is associated with high mortality rates, which are further exacerbated when accompanied by acute kidney injury (AKI). Presently, there is a lack of comprehensive studies thoroughly elucidating the metabolic dysregulation in ARDS patients with AKI leading to poor outcomes. We hypothesized that metabolomics can be a potent tool to highlight the differences in the metabolic profile unraveling unidentified pathophysiological mechanisms of ARDS patients with and without AKI. 1H nuclear magnetic resonance spectroscopy was used to identify key metabolites in the serum samples of 75 patients. Distinct clusters of both groups were obtained as the study's primary outcome using multivariate analysis. Notable alternations in the levels of nine metabolites were identified. Pathway analysis revealed the dysregulation of five significant cycles, which resulted in various complications, such as hyperammonemia, higher energy requirements, and mitochondrial dysfunction causing oxidative stress. Identified metabolites also showed a significant correlation with clinical scores, indicating severity. This study shows the alterations in the metabolite concentration highlighting the difference in the pathophysiology of both patient groups and its association with outcome, pointing in the direction of a personalized medicine approach and holding significant promise for application in critical care settings to improve clinical outcomes.
Collapse
Affiliation(s)
- Anamika Singh
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, India
- Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Mohd Adnan Siddiqui
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, India
| | - Swarnima Pandey
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, India
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 212001, United States
| | - Afzal Azim
- Department of Critical Care Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Neeraj Sinha
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, India
- Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
2
|
Lee M, Yoo JH, Kim I, Kang S, Lee W, Kim S, Han KS. The compartment-specific manipulation of the NAD +/NADH ratio affects the metabolome and the function of glioblastoma. Sci Rep 2024; 14:20575. [PMID: 39232046 PMCID: PMC11375122 DOI: 10.1038/s41598-024-71462-8] [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: 06/01/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of cancer in the brain and has an inferior prognosis because of the lack of suitable medicine, largely due to its tremendous invasion. GBM has selfish metabolic pathways to promote migration, invasion, and proliferation compared to normal cells. Among various metabolic pathways, NAD (nicotinamide adenine dinucleotide) is essential in generating ATP and is used as a resource for cancer cells. LbNOX (Lactobacillus brevis NADH oxidase) is an enzyme that can directly manipulate the NAD+/NADH ratio. In this study, we found that an increased NAD+/NADH ratio by LbNOX or mitoLbNOX reduced intracellular glutamate and calcium responses and reduced invasion capacity in GBM. However, the invasion was not affected in GBM by rotenone, an ETC (Electron Transport Chain) complex I inhibitor, or nicotinamide riboside, a NAD+ precursor, suggesting that the crucial factor is the NAD+/NADH ratio rather than the absolute quantity of ATP or NAD+ for the invasion of GBM. To develop a more accurate and effective GBM treatment, our findings highlight the importance of developing a new medicine that targets the regulation of the NAD+/NADH ratio, given the current lack of effective treatment options for this brain cancer.
Collapse
Affiliation(s)
- Myunghoon Lee
- Department of Biological Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Jae Hong Yoo
- Department of Biological Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Inseo Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sinbeom Kang
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Sungjin Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Kyung-Seok Han
- Department of Biological Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
3
|
Erb HHH, Polishchuk N, Stasyk O, Kahya U, Weigel MM, Dubrovska A. Glutamine Metabolism and Prostate Cancer. Cancers (Basel) 2024; 16:2871. [PMID: 39199642 PMCID: PMC11352381 DOI: 10.3390/cancers16162871] [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: 06/29/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Glutamine (Gln) is a non-essential amino acid that is involved in the development and progression of several malignancies, including prostate cancer (PCa). While Gln is non-essential for non-malignant prostate epithelial cells, PCa cells become highly dependent on an exogenous source of Gln. The Gln metabolism in PCa is tightly controlled by well-described oncogenes such as MYC, AR, and mTOR. These oncogenes contribute to therapy resistance and progression to the aggressive castration-resistant PCa. Inhibition of Gln catabolism impedes PCa growth, survival, and tumor-initiating potential while sensitizing the cells to radiotherapy. Therefore, given its significant role in tumor growth, targeting Gln metabolism is a promising approach for developing new therapeutic strategies. Ongoing clinical trials evaluate the safety and efficacy of Gln catabolism inhibitors in combination with conventional and targeted therapies in patients with various solid tumors, including PCa. Further understanding of how PCa cells metabolically interact with their microenvironment will facilitate the clinical translation of Gln inhibitors and help improve therapeutic outcomes. This review focuses on the role of Gln in PCa progression and therapy resistance and provides insights into current clinical trials.
Collapse
Affiliation(s)
- Holger H. H. Erb
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany;
| | - Nikita Polishchuk
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, 79000 Lviv, Ukraine; (N.P.); (O.S.)
| | - Oleh Stasyk
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, 79000 Lviv, Ukraine; (N.P.); (O.S.)
| | - Uğur Kahya
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (M.M.W.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
| | - Matthias M. Weigel
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (M.M.W.)
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (M.M.W.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01309 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| |
Collapse
|
4
|
Vallianatou T, Angerer TB, Kaya I, Nilsson A, Shariatgorji R, Svenningsson P, Andrén PE. Applying Spatial Metabolomics To Investigate Age- and Drug-Induced Neurochemical Changes. ACS Chem Neurosci 2024; 15:2822-2829. [PMID: 39072364 PMCID: PMC11311129 DOI: 10.1021/acschemneuro.4c00199] [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: 04/03/2024] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
In an era when population aging is increasing the burden of neurodegenerative conditions, deciphering the mechanisms underlying brain senescence is more important than ever. Here, we present a spatial metabolomics analysis of age-induced neurochemical alterations in the mouse brain using negative ionization mode mass spectrometry imaging. The age-dependent effects of the acetylcholinesterase inhibitor tacrine were simultaneously examined. For ultrahigh mass resolution analysis, we utilized a Fourier-transform ion cyclotron resonance spectrometer. To complement this, a trapped ion mobility spectrometry time-of-flight analyzer provided high speed and lateral resolution. The chosen approach facilitated the detection and identification of a wide range of metabolites, from amino acids to sphingolipids. We reported significant, age-dependent alterations in brain lipids which were most evident for sulfatides and lysophosphatidic acids. Sulfatide species, which are mainly localized to white matter, either increased or decreased with age, depending on the carbon chain length and hydroxylation stage. Lysophosphatidic acids were found to decrease with age in the detailed cortical and hippocampal subregions. An age-dependent increase in the glutamine/glutamate ratio, an indicator of glia-neuron interconnection and neurotoxicity, was detected after tacrine administration. The presented metabolic mapping approach was able to provide visualizations of the lipid signaling and neurotransmission alterations induced by early aging and can thus be beneficial to further elucidating age-related neurochemical pathways.
Collapse
Affiliation(s)
- Theodosia Vallianatou
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Tina B. Angerer
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Ibrahim Kaya
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Anna Nilsson
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Reza Shariatgorji
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Per Svenningsson
- Department
of Clinical Neuroscience, Karolinska Institute, Stockholm SE-17177, Sweden
| | - Per E. Andrén
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| |
Collapse
|
5
|
Chowdhury N, Wellslager B, Lee H, Gilbert JL, Yilmaz Ö. Glutamate is a key nutrient for Porphyromonas gingivalis growth and survival during intracellular autophagic life under nutritionally limited conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602514. [PMID: 39026746 PMCID: PMC11257440 DOI: 10.1101/2024.07.08.602514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Porphyromonas gingivalis survives in special autophagic vacuoles that serve as major replicative habitats in human primary gingival epithelial cells (GECs). As an asaccharolytic strict anaerobe, P. gingivalis is dependent on amino acids and peptides for nutrient sources. However, it is largely unknown as to P. gingivalis' metabolic processing under the nutritionally limited intracellular environments such the vacuoles, especially the preferred amino acids and associated-metabolic machineries. Here we elucidate that a Glutamate (Glu) catabolic enzyme, glutamate dehydrogenase (GdhA) is highly enriched in the isolated P. gingivalis -containing vacuoles. Interestingly, we found that P. gingivalis induces conversion of intracellular glutamine pool to Glu determined by analyses of the P. gingivalis- containing vacuoles and the whole infected-GECs. Critically, exogenous Glu-Glu dipeptide, a simple precursor of Glu, significantly increases the size of isolated intact P. gingivalis containing-vacuoles and live wild-type P. gingivalis numbers in GECs. In contrast, the isogenic GdhA-deficient-strain, Δ gdhA displayed a significant growth defect with collapsed-vacuoles in GECs. Next, we confirmed that P. gingivalis uptakes 14 C-Glu and it preferentially utilizes Glu-Glu-dipeptide using a nutritionally reduced Tryptic-Soy-Broth (TSB) media supplemented with Glu-Glu. Contrary, Δ gdhA -strain showed no detectable growth especially in nutritionally reduced TSB media with Glu-Glu. Using Atomic-Force-Microscopy, we observed that, wild-type P. gingivalis but not Δ gdhA strain notably increased the cell volume upon Glu-Glu supplementation, an indicator of higher metabolism and growth. Utilization of a human gingiva-mimicking organoid-system further validated the importance of Glu as an essential nutrient for the intramucosal colonization of P. gingivalis via the protected replicative vacuoles in GECs. Importance This study reveals that P. gingivalis heavily depends on preferential utilization of Glutamate (Glu) for autophagic vacuolar growth and survival in human GECs. Several novel observations are made to support this: (i) GdhA of P. gingivalis is highly enriched in these vacuoles, (ii) P. gingivalis induces a large conversion of intracellular glutamine to Glu, (iii) size of vacuoles are significantly increased in the presence of Glu-Glu in P. gingivalis wild-type strain infection which is opposite in a Δ gdhA strain, (iv) P. gingivalis uptakes 14 C-Glu and preferentially utilizes Glu-Glu dipeptide, (v) similarly, wild-type strain shows growth increase in a nutritionally reduced bacterial culture media, and (vi) finally, Glu-Glu supplementation increases bacterial cell-volume of P. gingivalis wild-type but not Δ gdhA strain, an indicator of higher metabolism and growth. Taken together, this study highlights the pathophysiological importance of Glu for P. gingivalis growth-rate, biomass induction and survival in nutritionally limited host subcellular environments.
Collapse
|
6
|
Lecoutre S, Maqdasy S, Rizo-Roca D, Renzi G, Vlassakev I, Alaeddine LM, Higos R, Jalkanen J, Zhong J, Zareifi DS, Frendo-Cumbo S, Massier L, Hodek O, Juvany M, Moritz T, de Castro Barbosa T, Omar-Hmeadi M, López-Yus M, Merabtene F, Abatan JB, Marcelin G, El Hachem EJ, Rouault C, Bergo MO, Petrus P, Zierath JR, Clément K, Krook A, Mejhert N, Rydén M. Reduced adipocyte glutaminase activity promotes energy expenditure and metabolic health. Nat Metab 2024; 6:1329-1346. [PMID: 39009762 PMCID: PMC11272588 DOI: 10.1038/s42255-024-01083-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/14/2024] [Indexed: 07/17/2024]
Abstract
Glutamine and glutamate are interconverted by several enzymes and alterations in this metabolic cycle are linked to cardiometabolic traits. Herein, we show that obesity-associated insulin resistance is characterized by decreased plasma and white adipose tissue glutamine-to-glutamate ratios. We couple these stoichiometric changes to perturbed fat cell glutaminase and glutamine synthase messenger RNA and protein abundance, which together promote glutaminolysis. In human white adipocytes, reductions in glutaminase activity promote aerobic glycolysis and mitochondrial oxidative capacity via increases in hypoxia-inducible factor 1α abundance, lactate levels and p38 mitogen-activated protein kinase signalling. Systemic glutaminase inhibition in male and female mice, or genetically in adipocytes of male mice, triggers the activation of thermogenic gene programs in inguinal adipocytes. Consequently, the knockout mice display higher energy expenditure and improved glucose tolerance compared to control littermates, even under high-fat diet conditions. Altogether, our findings highlight white adipocyte glutamine turnover as an important determinant of energy expenditure and metabolic health.
Collapse
Affiliation(s)
- Simon Lecoutre
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - David Rizo-Roca
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Gianluca Renzi
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Ivan Vlassakev
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Lynn M Alaeddine
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Romane Higos
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Jutta Jalkanen
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Jiawei Zhong
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Danae S Zareifi
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Scott Frendo-Cumbo
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Lucas Massier
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Ondrej Hodek
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Marta Juvany
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Thomas Moritz
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
- The Novo Nordisk Foundation Centre for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thais de Castro Barbosa
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Muhmmad Omar-Hmeadi
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Marta López-Yus
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, Zaragoza, Spain
- Instituto Aragonés de Ciencias de La Salud (IACS), Zaragoza, Spain
- Instituto de Investigación Sanitaria (IIS)-Aragón, Zaragoza, Spain
| | - Fatiha Merabtene
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Elie-Julien El Hachem
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Christine Rouault
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Martin O Bergo
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Paul Petrus
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Nutrition Department, Assistance Publique Hôpitaux de Paris, CRNH Ile-de-France, Pitié-Salpêtrière Hospital, Paris, France
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Niklas Mejhert
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden.
| | - Mikael Rydén
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden.
| |
Collapse
|
7
|
Martelli F, Quig A, Mele S, Lin J, Fulton TL, Wansbrough M, Barlow CK, Schittenhelm RB, Johnson TK, Piper MDW. A defined diet for pre-adult Drosophila melanogaster. Sci Rep 2024; 14:6974. [PMID: 38521863 PMCID: PMC10960813 DOI: 10.1038/s41598-024-57681-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/20/2024] [Indexed: 03/25/2024] Open
Abstract
Drosophila melanogaster is unique among animal models because it has a fully defined synthetic diet available to study nutrient-gene interactions. However, use of this diet is limited to adult studies due to impaired larval development and survival. Here, we provide an adjusted formula that reduces the developmental period, restores fat levels, enhances body mass, and fully rescues survivorship without compromise to adult lifespan. To demonstrate an application of this formula, we explored pre-adult diet compositions of therapeutic potential in a model of an inherited metabolic disorder affecting the metabolism of branched-chain amino acids. We reveal rapid, specific, and predictable nutrient effects on the disease state consistent with observations from mouse and patient studies. Together, our diet provides a powerful means with which to examine the interplay between diet and metabolism across all life stages in an animal model.
Collapse
Affiliation(s)
- Felipe Martelli
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Annelise Quig
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Sarah Mele
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Jiayi Lin
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Tahlia L Fulton
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Mia Wansbrough
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Christopher K Barlow
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Travis K Johnson
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia.
- Department of Biochemistry and Chemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia.
| | - Matthew D W Piper
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
8
|
Lackner S, Mahnert A, Moissl-Eichinger C, Madl T, Habisch H, Meier-Allard N, Kumpitsch C, Lahousen T, Kohlhammer-Dohr A, Mörkl S, Strobl H, Holasek S. Interindividual differences in aronia juice tolerability linked to gut microbiome and metabolome changes-secondary analysis of a randomized placebo-controlled parallel intervention trial. MICROBIOME 2024; 12:49. [PMID: 38461313 PMCID: PMC10924357 DOI: 10.1186/s40168-024-01774-4] [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: 07/11/2023] [Accepted: 02/05/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND Aronia melanocarpa is a berry rich in polyphenols known for health benefits. However, the bioavailability of polyphenols has been questioned, and the individual taste acceptance of the fruit with its specific flavor varies. We recently observed substantial differences in the tolerability of aronia juice among healthy females, with half of the individuals tolerating aronia juice without complaints. Given the importance of the gut microbiome in food digestion, we investigated in this secondary analysis of the randomized placebo-controlled parallel intervention study (ClinicalTrials.gov registration: NCT05432362) if aronia juice tolerability was associated with changes in intestinal microbiota and bacterial metabolites, seeking for potential mechanistic insights into the impact on aronia polyphenol tolerance and metabolic outcomes. RESULTS Forty females were enrolled for this 6-week trial, receiving either 100 ml natural aronia juice (verum, V) twice daily or a polyphenol-free placebo (P) with a similar nutritional profile, followed by a 6-week washout. Within V, individuals were categorized into those who tolerated the juice well (Vt) or reported complaints (Vc). The gut microbiome diversity, as analyzed by 16S rRNA gene-based next-generation sequencing, remained unaltered in Vc but changed significantly in Vt. A MICOM-based flux balance analysis revealed pronounced differences in the 40 most predictive metabolites post-intervention. In Vc carbon-dioxide, ammonium and nine O-glycans were predicted due to a shift in microbial composition, while in Vt six bile acids were the most likely microbiota-derived metabolites. NMR metabolomics of plasma confirmed increased lipoprotein subclasses (LDL, VLDL) post-intervention, reverting after wash out. Stool samples maintained a stable metabolic profile. CONCLUSION In linking aronia polyphenol tolerance to gut microbiota-derived metabolites, our study explores adaptive processes affecting lipoprotein profiles during high polyphenol ingestion in Vt and examines effects on mucosal gut health in response to intolerance to high polyphenol intake in Vc. Our results underpin the importance of individualized hormetic dosing for beneficial polyphenol effects, demonstrate dynamic gut microbiome responses to aronia juice, and emphasize personalized responses in polyphenol interventions.
Collapse
Affiliation(s)
- Sonja Lackner
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Stiftingtalstraße 6, 8010, Graz, Austria
| | - Alexander Mahnert
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Christine Moissl-Eichinger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010, Graz, Austria
| | - Tobias Madl
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010, Graz, Austria
| | - Hansjörg Habisch
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Nathalie Meier-Allard
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Stiftingtalstraße 6, 8010, Graz, Austria
| | - Christina Kumpitsch
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Theresa Lahousen
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Auenbruggerplatz 31, 8036, Graz, Austria
| | - Alexandra Kohlhammer-Dohr
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Auenbruggerplatz 31, 8036, Graz, Austria
| | - Sabrina Mörkl
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Auenbruggerplatz 3, 8036, Graz, Austria
| | - Herbert Strobl
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Stiftingtalstraße 6, 8010, Graz, Austria
| | - Sandra Holasek
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Stiftingtalstraße 6, 8010, Graz, Austria.
| |
Collapse
|
9
|
Mhanna A, Martini N, Hmaydoosh G, Hamwi G, Jarjanazi M, Zaifah G, Kazzazo R, Haji Mohamad A, Alshehabi Z. The correlation between gut microbiota and both neurotransmitters and mental disorders: A narrative review. Medicine (Baltimore) 2024; 103:e37114. [PMID: 38306525 PMCID: PMC10843545 DOI: 10.1097/md.0000000000037114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/09/2024] [Indexed: 02/04/2024] Open
Abstract
The gastrointestinal tract is embedded with microorganisms of numerous genera, referred to as gut microbiota. Gut microbiota has multiple effects on many body organs, including the brain. There is a bidirectional connection between the gut and brain called the gut-brain-axis, and these connections are formed through immunological, neuronal, and neuroendocrine pathways. In addition, gut microbiota modulates the synthesis and functioning of neurotransmitters. Therefore, the disruption of the gut microbiota in the composition or function, which is known as dysbiosis, is associated with the pathogenesis of many mental disorders, such as schizophrenia, depression, and other psychiatric disorders. This review aims to summarize the modulation role of the gut microbiota in 4 prominent neurotransmitters (tryptophan and serotonergic system, dopamine, gamma-aminobutyric acid, and glutamate), as well as its association with 4 psychiatric disorders (schizophrenia, depression, anxiety disorders, and autism spectrum disorder). More future research is required to develop efficient gut-microbiota-based therapies for these illnesses.
Collapse
Affiliation(s)
- Amjad Mhanna
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Nafiza Martini
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
- Damascus University, Faculty of Medicine, Damascus, Syrian Arab Republic
| | - Ghefar Hmaydoosh
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - George Hamwi
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Mulham Jarjanazi
- Pediatric Surgery Resident, Pediatric Surgery Department, Aleppo University Hospital, Aleppo, Syrian Arab Republic
| | - Ghaith Zaifah
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Reem Kazzazo
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Aya Haji Mohamad
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
- Faculty of Medicine, Aleppo University, Aleppo University Hospital, Aleppo, Syrian Arab Republic
| | - Zuheir Alshehabi
- Department of Pathology, Tishreen University Hospital, Latakia, Syrian Arab Republic
| |
Collapse
|
10
|
Viudez-Martínez A, Torregrosa AB, Navarrete F, García-Gutiérrez MS. Understanding the Biological Relationship between Migraine and Depression. Biomolecules 2024; 14:163. [PMID: 38397400 PMCID: PMC10886628 DOI: 10.3390/biom14020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
Migraine is a highly prevalent neurological disorder. Among the risk factors identified, psychiatric comorbidities, such as depression, seem to play an important role in its onset and clinical course. Patients with migraine are 2.5 times more likely to develop a depressive disorder; this risk becomes even higher in patients suffering from chronic migraine or migraine with aura. This relationship is bidirectional, since depression also predicts an earlier/worse onset of migraine, increasing the risk of migraine chronicity and, consequently, requiring a higher healthcare expenditure compared to migraine alone. All these data suggest that migraine and depression may share overlapping biological mechanisms. Herein, this review explores this topic in further detail: firstly, by introducing the common epidemiological and risk factors for this comorbidity; secondly, by focusing on providing the cumulative evidence of common biological aspects, with a particular emphasis on the serotoninergic system, neuropeptides such as calcitonin-gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), substance P, neuropeptide Y and orexins, sexual hormones, and the immune system; lastly, by remarking on the future challenges required to elucidate the etiopathological mechanisms of migraine and depression and providing updated information regarding new key targets for the pharmacological treatment of these clinical entities.
Collapse
Affiliation(s)
- Adrián Viudez-Martínez
- Hospital Pharmacy Service, Hospital General Dr. Balmis de Alicante, 03010 Alicante, Spain;
| | - Abraham B. Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
11
|
Saha B, Barman S, Majumder S, Ghosh B, Mallick K, Choudhury S, Roy MN. Investigation of intermolecular interactions of l-Valine and l-Phenylalanine with muscle relaxant drug mephenesin molecule prevalent in aqueous solution by various physico-chemical methods at T=298.15K-313.15K. Heliyon 2024; 10:e23562. [PMID: 38173535 PMCID: PMC10761785 DOI: 10.1016/j.heliyon.2023.e23562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
A complete chemical analysis of significant intermolecular interactions of l-Valine (L-Val) and l-Phenylalanine (L-Phe) with Mephenesin (MEPN) molecules in aqueous solution has been studied by different physicochemical methodologies at various temperatures (T = 298.15 K-313.15 K at an interval of 5 K) and concentrations (0.001 mol kg-1, 0.003 mol kg-1, 0.005 mol kg-1) of aqueous MEPN solution. The limiting apparent molar volume (φ V 0 ) and experimental slope (S V * ) values are found from the equation of Masson, viscosity A and B-coefficient determined using the equation of Jones-Doles, molar refraction (RM) and limiting molar refraction (RM0) derived by the Lorentz-Lorenz equation, express that in our experimental solution of amino acids (AAs) in aqueous MEPN, the solute-solvent interaction predominates over the solute-solute and solvent-solvent interactions for these ternary solutions. These are also justified by the measurement of various thermodynamic parameters, free energy of activation of viscous flow per mole of solvent(Δμ1°#) and solute (Δμ2°#), activation of viscous flow of enthalpies (ΔH°#) and entropies (ΔS°#). The characteristics of structure-breaking of solutes in the aqueous drug solution have been identified by Hepler's method and dB/dT value. The spectroscopic methods like UV-visible and proton-NMR studies help to explicate the strong AA-MEPN interactions in the solution phase and obtain a good correlation with theoretical studies. Theoretical investigations are checked to authenticate the experimental observations and according to both studies, L-Phe-MEPN interaction is greater than L-Val-MEPN interaction. The experimental and correlated research data are useful for the development of model combinations of AAs with drug molecules in pharmaceutical and medicinal chemistry.
Collapse
Affiliation(s)
- Baishali Saha
- Department of Chemistry, University of North Bengal, Darjeeling, 734013, India
| | - Sanjoy Barman
- Department of Chemistry, University of North Bengal, Darjeeling, 734013, India
| | - Sukdev Majumder
- Department of Chemistry, University of North Bengal, Darjeeling, 734013, India
| | - Biswajit Ghosh
- Department of Chemistry, University of North Bengal, Darjeeling, 734013, India
| | - Kangkan Mallick
- Department of Chemistry, University of North Bengal, Darjeeling, 734013, India
| | | | - Mahendra Nath Roy
- Department of Chemistry, University of North Bengal, Darjeeling, 734013, India
- Vice-Chancellor, Alipurduar University, Alipurduar 736121, 734013, Darjeeling, India
| |
Collapse
|
12
|
Rong H, Wang D, Wang Y, Dong C, Wang G. YTHDF1 in Tumor Cell Metabolism: An Updated Review. Molecules 2023; 29:140. [PMID: 38202722 PMCID: PMC10779796 DOI: 10.3390/molecules29010140] [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: 11/22/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
With the advancement of research on m6A-related mechanisms in recent years, the YTHDF protein family within m6A readers has garnered significant attention. Among them, YTHDF1 serves as a pivotal member, playing a crucial role in protein translation, tumor proliferation, metabolic reprogramming of various tumor cells, and immune evasion. In addition, YTHDF1 also exerts regulatory effects on tumors through multiple signaling pathways, and numerous studies have confirmed its ability to assist in the reprogramming of the tumor cell-related metabolic processes. The focus of research on YTHDF1 has shifted in recent years from its m6A-recognition and -modification function to the molecular mechanisms by which it regulates tumor progression, particularly by exploring the regulatory factors that interact with YTHDF1 upstream and downstream. In this review, we elucidate the latest signaling pathway mechanisms of YTHDF1 in various tumor cells, with a special emphasis on its distinctive characteristics in tumor cell metabolic reprogramming. Furthermore, we summarize the latest pathological and physiological processes involving YTHDF1 in tumor cells, and analyze potential therapeutic approaches that utilize YTHDF1. We believe that YTHDF1 represents a highly promising target for future tumor treatments and a novel tumor biomarker.
Collapse
Affiliation(s)
| | | | | | | | - Guiling Wang
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, China; (H.R.); (D.W.); (Y.W.); (C.D.)
| |
Collapse
|
13
|
Baranovicova E, Kalenska D, Kaplan P, Kovalska M, Tatarkova Z, Lehotsky J. Blood and Brain Metabolites after Cerebral Ischemia. Int J Mol Sci 2023; 24:17302. [PMID: 38139131 PMCID: PMC10743907 DOI: 10.3390/ijms242417302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
The study of an organism's response to cerebral ischemia at different levels is essential to understanding the mechanism of the injury and protection. A great interest is devoted to finding the links between quantitative metabolic changes and post-ischemic damage. This work aims to summarize the outcomes of the most studied metabolites in brain tissue-lactate, glutamine, GABA (4-aminobutyric acid), glutamate, and NAA (N-acetyl aspartate)-regarding their biological function in physiological conditions and their role after cerebral ischemia/reperfusion. We focused on ischemic damage and post-ischemic recovery in both experimental-including our results-as well as clinical studies. We discuss the role of blood glucose in view of the diverse impact of hyperglycemia, whether experimentally induced, caused by insulin resistance, or developed as a stress response to the cerebral ischemic event. Additionally, based on our and other studies, we analyze and critically discuss post-ischemic alterations in energy metabolites and the elevation of blood ketone bodies observed in the studies on rodents. To complete the schema, we discuss alterations in blood plasma circulating amino acids after cerebral ischemia. So far, no fundamental brain or blood metabolite(s) has been recognized as a relevant biological marker with the feasibility to determine the post-ischemic outcome or extent of ischemic damage. However, studies from our group on rats subjected to protective ischemic preconditioning showed that these animals did not develop post-ischemic hyperglycemia and manifested a decreased metabolic infringement and faster metabolomic recovery. The metabolomic approach is an additional tool for understanding damaging and/or restorative processes within the affected brain region reflected in the blood to uncover the response of the whole organism via interorgan metabolic communications to the stressful cerebral ischemic challenge.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia;
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia
| | - Peter Kaplan
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia
| | - Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| |
Collapse
|
14
|
Yang Y, Han Z, Gao Z, Chen J, Song C, Xu J, Wang H, Huang A, Shi J, Gu J. Metagenomic and targeted metabolomic analyses reveal distinct phenotypes of the gut microbiota in patients with colorectal cancer and type 2 diabetes mellitus. Chin Med J (Engl) 2023; 136:2847-2856. [PMID: 36959686 PMCID: PMC10686596 DOI: 10.1097/cm9.0000000000002421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is an independent risk factor for colorectal cancer (CRC), and the patients with CRC and T2DM have worse survival. The human gut microbiota (GM) is linked to the development of CRC and T2DM, respectively. However, the GM characteristics in patients with CRC and T2DM remain unclear. METHODS We performed fecal metagenomic and targeted metabolomics studies on 36 samples from CRC patients with T2DM (DCRC group, n = 12), CRC patients without diabetes (CRC group, n = 12), and healthy controls (Health group, n = 12). We analyzed the fecal microbiomes, characterized the composition and function based on the metagenomics of DCRC patients, and detected the short-chain fatty acids (SCFAs) and bile acids (BAs) levels in all fecal samples. Finally, we performed a correlation analysis of the differential bacteria and metabolites between different groups. RESULTS Compared with the CRC group, LefSe analysis showed that there is a specific GM community in DCRC group, including an increased abundance of Eggerthella , Hungatella , Peptostreptococcus , and Parvimonas , and decreased Butyricicoccus , Lactobacillus , and Paraprevotella . The metabolomics analysis results revealed that the butyric acid level was lower but the deoxycholic acid and 12-keto-lithocholic acid levels were higher in the DCRC group than other groups ( P < 0.05). The correlation analysis showed that the dominant bacterial abundance in the DCRC group ( Parvimonas , Desulfurispora , Sebaldella , and Veillonellales , among others) was negatively correlated with butyric acid, hyodeoxycholic acid, ursodeoxycholic acid, glycochenodeoxycholic acid, chenodeoxycholic acid, cholic acid and glycocholate. However, the abundance of mostly inferior bacteria was positively correlated with these metabolic acid levels, including Faecalibacterium , Thermococci , and Cellulophaga . CONCLUSIONS Unique fecal microbiome signatures exist in CRC patients with T2DM compared to those with non-diabetic CRC. Alterations in GM composition and SCFAs and secondary BAs levels may promote CRC development.
Collapse
Affiliation(s)
- Yong Yang
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing 100144, China
| | - Zihan Han
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Zhaoya Gao
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing 100144, China
| | - Jiajia Chen
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing 100144, China
| | - Can Song
- Peking-Tsinghua Center for Life Science, Peking University International Cancer Center, Beijing 100142, China
| | - Jingxuan Xu
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Hanyang Wang
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - An Huang
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jingyi Shi
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jin Gu
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing 100144, China
- Peking-Tsinghua Center for Life Science, Peking University International Cancer Center, Beijing 100142, China
| |
Collapse
|
15
|
Zhang X, Wang H, Kilpatrick LA, Dong TS, Gee GC, Labus JS, Osadchiy V, Beltran-Sanchez H, Wang MC, Vaughan A, Gupta A. Discrimination exposure impacts unhealthy processing of food cues: crosstalk between the brain and gut. NATURE MENTAL HEALTH 2023; 1:841-852. [PMID: 38094040 PMCID: PMC10718506 DOI: 10.1038/s44220-023-00134-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/28/2023] [Indexed: 12/17/2023]
Abstract
Experiences of discrimination are associated with adverse health outcomes, including obesity. However, the mechanisms by which discrimination leads to obesity remain unclear. Utilizing multi-omics analyses of neuroimaging and fecal metabolites, we investigated the impact of discrimination exposure on brain reactivity to food images and associated dysregulations in the brain-gut-microbiome system. We show that discrimination is associated with increased food-cue reactivity in frontal-striatal regions involved in reward, motivation and executive control; altered glutamate-pathway metabolites involved in oxidative stress and inflammation as well as preference for unhealthy foods. Associations between discrimination-related brain and gut signatures were skewed towards unhealthy sweet foods after adjusting for age, diet, body mass index, race and socioeconomic status. Discrimination, as a stressor, may contribute to enhanced food-cue reactivity and brain-gut-microbiome disruptions that can promote unhealthy eating behaviors, leading to increased risk for obesity. Treatments that normalize these alterations may benefit individuals who experience discrimination-related stress.
Collapse
Affiliation(s)
- Xiaobei Zhang
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Hao Wang
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA, USA
- School of Physics and Optoelectronic Engineering, Hainan University, Haikou, China
| | - Lisa A. Kilpatrick
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Tien S. Dong
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Goodman–Luskin Microbiome Center, UCLA, Los Angeles, CA, USA
| | - Gilbert C. Gee
- University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Community Health Sciences Fielding School of Public Health, UCLA, Los Angeles, CA, USA
- California Center for Population Research, UCLA, Los Angeles, CA, USA
| | - Jennifer S. Labus
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Goodman–Luskin Microbiome Center, UCLA, Los Angeles, CA, USA
| | - Vadim Osadchiy
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Urology, UCLA, Los Angeles, CA, USA
| | - Hiram Beltran-Sanchez
- University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Community Health Sciences Fielding School of Public Health, UCLA, Los Angeles, CA, USA
- California Center for Population Research, UCLA, Los Angeles, CA, USA
| | - May C. Wang
- University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Community Health Sciences Fielding School of Public Health, UCLA, Los Angeles, CA, USA
| | - Allison Vaughan
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Goodman–Luskin Microbiome Center, UCLA, Los Angeles, CA, USA
| |
Collapse
|
16
|
Zhou X, Zhang J, Sun Y, Shen J, Sun B, Ma Q. Glutamine Ameliorates Liver Steatosis via Regulation of Glycolipid Metabolism and Gut Microbiota in High-Fat Diet-Induced Obese Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15656-15667. [PMID: 37847053 DOI: 10.1021/acs.jafc.3c05566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Obesity and its associated conditions, such as nonalcoholic fatty liver disease (NAFLD), are risk factors for health. The aim of this study was to explore the effects of glutamine (Gln) on liver steatosis induced by a high-fat diet (HFD) and HEPG2 cells induced by oleic acid. Gln demonstrated a positive influence on hepatic homeostasis by suppressing acetyl CoA carboxylase (ACC) and fatty acid synthase (FAS) and promoting sirtuin 1 (SIRT1) expression while improving glucose metabolism by regulating serine/threonine protein kinase (AKT)/factor forkhead box O1 (FOXO1) signals in vivo and in vitro. Obese Gln-fed mice had higher colonic short-chain fatty acid (SCFA) contents and lower inflammation factor protein levels in the liver, HEPG2 cells, and jejunum. Gln-treated obese mice had an effective decrease in Firmicutes abundance. These findings indicate that Gln serves as a nutritional tool in managing obesity and related disorders.
Collapse
Affiliation(s)
- Xinbo Zhou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Junjie Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yutong Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Jian Shen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Bo Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Qingquan Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
17
|
Sánchez A, García-Pardo G, Gómez-Bertomeu F, López-Dupla M, Foguet-Romero E, Buzón MJ, Almirante B, Olona M, Fernández-Veledo S, Vidal F, Chafino S, Rull A, Peraire J. Mitochondrial dysfunction, lipids metabolism, and amino acid biosynthesis are key pathways for COVID-19 recovery. iScience 2023; 26:107948. [PMID: 37810253 PMCID: PMC10551651 DOI: 10.1016/j.isci.2023.107948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/29/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023] Open
Abstract
The metabolic alterations caused by SARS-CoV-2 infection reflect disease progression. To analyze molecules involved in these metabolic changes, a multiomics study was performed using plasma from 103 patients with different degrees of COVID-19 severity during the evolution of the infection. With the increased severity of COVID-19, changes in circulating proteomic, metabolomic, and lipidomic profiles increased. Notably, the group of severe and critical patients with high HRG and ChoE (20:3) and low alpha-ketoglutaric acid levels had a high chance of unfavorable disease evolution (AUC = 0.925). Consequently, patients with the worst prognosis presented alterations in the TCA cycle (mitochondrial dysfunction), lipid metabolism, amino acid biosynthesis, and coagulation. Our findings increase knowledge regarding how SARS-CoV-2 infection affects different metabolic pathways and help in understanding the future consequences of COVID-19 to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Alba Sánchez
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Graciano García-Pardo
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Fréderic Gómez-Bertomeu
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Miguel López-Dupla
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Elisabet Foguet-Romero
- Eurecat, Centre Tecnologic de Catalunya, Centre for Omic Sciences (Joint Unit Eurecat - Universitat Rovira i Virgili), Unique Scientific and Technical Infrastructure (ICTS), Reus, Spain
| | - Maria José Buzón
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Infectious Diseases Department, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, (VHIR) Task Force COVID-19, Barcelona, Spain
| | - Benito Almirante
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Montserrat Olona
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Sonia Fernández-Veledo
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Francesc Vidal
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Silvia Chafino
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Anna Rull
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Joaquim Peraire
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - for the COVIDOMICS Study Group
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Eurecat, Centre Tecnologic de Catalunya, Centre for Omic Sciences (Joint Unit Eurecat - Universitat Rovira i Virgili), Unique Scientific and Technical Infrastructure (ICTS), Reus, Spain
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Infectious Diseases Department, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, (VHIR) Task Force COVID-19, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
18
|
Di Maio A, Nuzzo T, Gilio L, Serra M, Buttari F, Errico F, De Rosa A, Bassi MS, Morelli M, Sasabe J, Sulzer D, Carta M, Centonze D, Usiello A. Homeostasis of serine enantiomers is disrupted in the post-mortem caudate putamen and cerebrospinal fluid of living Parkinson's disease patients. Neurobiol Dis 2023:106203. [PMID: 37336364 DOI: 10.1016/j.nbd.2023.106203] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023] Open
Abstract
L-serine generated in astrocytes plays a pivotal role in modulating essential neurometabolic processes, while its enantiomer, D-serine, specifically regulates NMDA receptor (NMDAR) signalling. Despite their physiological relevance in modulating cerebral activity, serine enantiomers metabolism in Parkinson's disease (PD) remains elusive. Using High-Performance Liquid Chromatography (HPLC), we measured D- and L-serine levels along with other amino acids known to modulate NMDAR function, such as L-glutamate, L-aspartate, D-aspartate, and glycine, in the post-mortem caudate putamen (CPu) and superior frontal gyrus (SFG) of PD patients. Moreover, we examined these amino acids in the cerebrospinal fluid (CSF) of de novo living PD, Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS) patients versus subjects with other neurological disorders (OND), used as control. We found higher D-serine and L-serine levels in the CPu of PD patients but not in the SFG, a cerebral region that, in contrast to the CPu, is not innervated by nigral dopaminergic terminals. We also highlighted a significant elevation of both serine enantiomers in the CSF samples from PD but not in those of AD and ALS patients, compared with control subjects. By contrast, none or only minor changes were found in the amount of other neuroactive amino acids mentioned above. Our findings identify D-serine and L-serine level upregulation as a biochemical signature associated with nigrostriatal dopaminergic degeneration in PD.
Collapse
Affiliation(s)
- Anna Di Maio
- Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Tommaso Nuzzo
- Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Luana Gilio
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy; Faculty of Psychology, Uninettuno Telematic International University, Rome, Italy; Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy
| | - Marcello Serra
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Francesco Errico
- Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Department of Agricultural Sciences, University of Naples "Federico II", Portici, Italy
| | - Arianna De Rosa
- Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | | | - Micaela Morelli
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy; National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Jumpei Sasabe
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - David Sulzer
- Departments of Psychiatry, Neurology, Pharmacology, Columbia University Irving Medical Center, Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Diego Centonze
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Alessandro Usiello
- Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy.
| |
Collapse
|
19
|
Kim M, Oh S, Kim S, Ji M, Choi B, Bae JW, Lee YS, Paik MJ, Lee S. Alcohol perturbed locomotor behavior, metabolism, and pharmacokinetics of gamma-hydroxybutyric acid in rats. Biomed Pharmacother 2023; 164:114992. [PMID: 37301134 DOI: 10.1016/j.biopha.2023.114992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/12/2023] Open
Abstract
Gamma-hydroxybutyric acid (GHB), both a metabolic precursor and product of gamma-aminobutyric acid (GABA), is a central nervous system depressant used for the treatment of narcolepsy-associated cataplexy and alcohol withdrawal. However, administration of GHB with alcohol (ethanol) is a major cause of hospitalizations related to GHB intoxication. In this study, we investigated locomotor behavior as well as metabolic and pharmacokinetic interactions following co-administration of GHB and ethanol in rats. The locomotor behavior of rats was evaluated following the intraperitoneal administration of GHB (sodium salt, 500 mg/kg) and/or ethanol (2 g/kg). Further, time-course urinary metabolic profiling of GHB and its biomarker metabolites glutamic acid, GABA, succinic acid, 2,4-dihydroxybutyric acid (OH-BA), 3,4-OH-BA, and glycolic acid as well as pharmacokinetic analysis were performed. GHB/ethanol co-administration significantly reduced locomotor activity, compared to the individual administration of GHB or ethanol. The urinary and plasma concentrations of GHB and other target compounds, except for 2,4-OH-BA, were significantly higher in the GHB/ethanol co-administration group than the group administered only GHB. The pharmacokinetic analysis results showed that the co-administration of GHB and ethanol significantly increased the half-life of GHB while the total clearance decreased. Moreover, a comparison of the metabolite-to-parent drug area under the curve ratios demonstrated that the metabolic pathways of GHB, such α- and β-oxidation, were inhibited by ethanol. Consequently, the co-administration of GHB and ethanol aggravated the metabolism and elimination of GHB and enhanced its sedative effect. These findings will contribute to clinical interpretation of GHB intoxication.
Collapse
Affiliation(s)
- Mingyu Kim
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, the Republic of Korea
| | - Songjin Oh
- College of Pharmacy, Sunchon National University, 25 Jungang-ro, Suncheon 57922, the Republic of Korea
| | - Suji Kim
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, the Republic of Korea
| | - Moongi Ji
- College of Pharmacy, Sunchon National University, 25 Jungang-ro, Suncheon 57922, the Republic of Korea
| | - Byeongchan Choi
- College of Pharmacy, Sunchon National University, 25 Jungang-ro, Suncheon 57922, the Republic of Korea
| | - Jung-Woo Bae
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, the Republic of Korea
| | - Yong Sup Lee
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, the Republic of Korea
| | - Man-Jeong Paik
- College of Pharmacy, Sunchon National University, 25 Jungang-ro, Suncheon 57922, the Republic of Korea.
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, the Republic of Korea.
| |
Collapse
|
20
|
Zhu S, Li Y, Zhang F, Xiong C, Gao H, Yao Y, Qian W, Ding C, Chen S. Raman spectromics method for fast and label-free genotype screening. BIOMEDICAL OPTICS EXPRESS 2023; 14:3072-3085. [PMID: 37342689 PMCID: PMC10278603 DOI: 10.1364/boe.493524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/23/2023]
Abstract
It is now understood that genes and their various mutations are associated with the onset and progression of diseases. However, routine genetic testing techniques are limited by their high cost, time consumption, susceptibility to contamination, complex operation, and data analysis difficulties, rendering them unsuitable for genotype screening in many cases. Therefore, there is an urgent need to develop a rapid, sensitive, user-friendly, and cost-effective method for genotype screening and analysis. In this study, we propose and investigate a Raman spectroscopic method for achieving fast and label-free genotype screening. The method was validated using spontaneous Raman measurements of wild-type Cryptococcus neoformans and its six mutants. An accurate identification of different genotypes was achieved by employing a one-dimensional convolutional neural network (1D-CNN), and significant correlations between metabolic changes and genotypic variations were revealed. Genotype-specific regions of interest were also localized and visualized using a gradient-weighted class activation mapping (Grad-CAM)-based spectral interpretable analysis method. Furthermore, the contribution of each metabolite to the final genotypic decision-making was quantified. The proposed Raman spectroscopic method demonstrated huge potential for fast and label-free genotype screening and analysis of conditioned pathogens.
Collapse
Affiliation(s)
- Shanshan Zhu
- Research Institute of Medical and Biological Engineering, Ningbo University, Ningbo 315211, China
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
- Health Science Center, Ningbo University, Ningbo 315211, China
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou 350117, China
| | - Yanjian Li
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Fengdi Zhang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
| | - Changchun Xiong
- College of Electrical Engineering and Computer Science, Ningbo University, Ningbo 315211, China
| | - Han Gao
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
| | - Yudong Yao
- Research Institute of Medical and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Wei Qian
- Research Institute of Medical and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Chen Ding
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Shuo Chen
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Shenyang 110169, China
| |
Collapse
|
21
|
Pérez-Sala D, Pajares MA. Appraising the Role of Astrocytes as Suppliers of Neuronal Glutathione Precursors. Int J Mol Sci 2023; 24:ijms24098059. [PMID: 37175763 PMCID: PMC10179008 DOI: 10.3390/ijms24098059] [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: 03/15/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The metabolism and intercellular transfer of glutathione or its precursors may play an important role in cellular defense against oxidative stress, a common hallmark of neurodegeneration. In the 1990s, several studies in the Neurobiology field led to the widely accepted notion that astrocytes produce large amounts of glutathione that serve to feed neurons with precursors for glutathione synthesis. This assumption has important implications for health and disease since a reduction in this supply from astrocytes could compromise the capacity of neurons to cope with oxidative stress. However, at first glance, this shuttling would imply a large energy expenditure to get to the same point in a nearby cell. Thus, are there additional underlying reasons for this expensive mechanism? Are neurons unable to import and/or synthesize the three non-essential amino acids that are the glutathione building blocks? The rather oxidizing extracellular environment favors the presence of cysteine (Cys) as cystine (Cis), less favorable for neuronal import. Therefore, it has also been proposed that astrocytic GSH efflux could induce a change in the redox status of the extracellular space nearby the neurons, locally lowering the Cis/Cys ratio. This astrocytic glutathione release would also increase their demand for precursors, stimulating Cis uptake, which these cells can import, further impacting the local decline of the Cis/Cys ratio, in turn, contributing to a more reduced extracellular environment and subsequently favoring neuronal Cys import. Here, we revisit the experimental evidence that led to the accepted hypothesis of astrocytes acting as suppliers of neuronal glutathione precursors, considering recent data from the Human Protein Atlas. In addition, we highlight some potential drawbacks of this hypothesis, mainly supported by heterogeneous cellular models. Finally, we outline additional and more cost-efficient possibilities by which astrocytes could support neuronal glutathione levels, including its shuttling in extracellular vesicles.
Collapse
Affiliation(s)
- Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - María A Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| |
Collapse
|
22
|
Ren S, Zhang Z, Song Q, Ren Z, Xiao J, Li L, Zhang Q. Metabolic exploration of the developmental abnormalities and neurotoxicity of Esculentoside B, the main toxic factor in Phytolaccae radix. Food Chem Toxicol 2023; 176:113777. [PMID: 37080526 DOI: 10.1016/j.fct.2023.113777] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 04/22/2023]
Abstract
P: radix is a perennial herb, and its extracts have various biological properties that make it a potential candidate for the treatment of tumors, edema, and lymphatic stasis. However, the main factor contributing to its toxicity are not clear. Here, we used a zebrafish toxicological model to study the main toxicity factor of P. radix and explore the potential mechanisms involved. The results revealed that Esculentoside B was the major toxic factor of P. radix. Exposure of zebrafish larvae to Esculentoside B caused developmental abnormalities, neurotoxicity and altered locomotor behavior. The combination of AChE activity and the expression levels of genes relevant to CNS development demonstrated that Esculentoside B is neurotoxic to zebrafish larvae, impairs their CNS development, and that AChE may be a toxic target of Esculentoside B. Metabolomic analysis has revealed that Esculentoside B exposure can disrupt D-Amino acid metabolism, protein export, autophagy, and mTOR signaling pathways in zebrafish larvae. These findings provide insights into the molecular mechanisms underlying EsB-induced neurotoxicity in zebrafish, which can facilitate further research and development of P. radix for safe consumption.
Collapse
Affiliation(s)
- Sipei Ren
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shanxi, China
| | - Zhichao Zhang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shanxi, China
| | - Qinyang Song
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shanxi, China
| | - Zhaoyang Ren
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shanxi, China
| | - Jian Xiao
- Shaanxi Key Laboratory of Phytochemistry, College of Chemistry and Chemical Engineering, Baoji University of Arts and Sciences, Baoji, 721013, China.
| | - Luqi Li
- Life Science Research Core Services, Northwest A&F University, Yangling, Xianyang, 712100, China
| | - Qiang Zhang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shanxi, China; Shaanxi Key Laboratory of Phytochemistry, College of Chemistry and Chemical Engineering, Baoji University of Arts and Sciences, Baoji, 721013, China.
| |
Collapse
|
23
|
Jin J, Byun JK, Choi YK, Park KG. Targeting glutamine metabolism as a therapeutic strategy for cancer. Exp Mol Med 2023; 55:706-715. [PMID: 37009798 PMCID: PMC10167356 DOI: 10.1038/s12276-023-00971-9] [Citation(s) in RCA: 111] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 04/04/2023] Open
Abstract
Proliferating cancer cells rely largely on glutamine for survival and proliferation. Glutamine serves as a carbon source for the synthesis of lipids and metabolites via the TCA cycle, as well as a source of nitrogen for amino acid and nucleotide synthesis. To date, many studies have explored the role of glutamine metabolism in cancer, thereby providing a scientific rationale for targeting glutamine metabolism for cancer treatment. In this review, we summarize the mechanism(s) involved at each step of glutamine metabolism, from glutamine transporters to redox homeostasis, and highlight areas that can be exploited for clinical cancer treatment. Furthermore, we discuss the mechanisms underlying cancer cell resistance to agents that target glutamine metabolism, as well as strategies for overcoming these mechanisms. Finally, we discuss the effects of glutamine blockade on the tumor microenvironment and explore strategies to maximize the utility of glutamine blockers as a cancer treatment.
Collapse
Affiliation(s)
- Jonghwa Jin
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Jun-Kyu Byun
- BK21 FOUR Community-based Intelligent Novel Drug Discovery Education Unit, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, 41566, Korea
| | - Yeon-Kyung Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, 41404, Korea.
| | - Keun-Gyu Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea.
| |
Collapse
|
24
|
Boyko M, Gruenbaum BF, Frank D, Natanel D, Negev S, Azab AN, Barsky G, Knyazer B, Kofman O, Zlotnik A. The Integrity of the Blood-Brain Barrier as a Critical Factor for Regulating Glutamate Levels in Traumatic Brain Injury. Int J Mol Sci 2023; 24:ijms24065897. [PMID: 36982969 PMCID: PMC10056818 DOI: 10.3390/ijms24065897] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
A healthy blood-brain barrier (BBB) shields the brain from high concentrations of blood glutamate, which can cause neurotoxicity and neurodegeneration. It is believed that traumatic brain injury (TBI) causes long-term BBB disruption, subsequently increasing brain glutamate in the blood, in addition to increased glutamate resulting from the neuronal injury. Here, we investigate the relationship between blood and brain glutamate levels in the context of BBB permeability. Rats exposed to BBB disruption through an osmotic model or TBI and treated with intravenous glutamate or saline were compared to control rats with an intact BBB treated with intravenous glutamate or saline. After BBB disruption and glutamate administration, the concentrations of glutamate in the cerebrospinal fluid and blood and brain tissue were analyzed. The results showed a strong correlation between the brain and blood glutamate concentrations in the groups with BBB disruption. We conclude that a healthy BBB protects the brain from high levels of blood glutamate, and the permeability of the BBB is a vital component in regulating levels of glutamate in the brain. These findings bring a new approach to treating the consequences of TBI and other diseases where long-term disruption of the BBB is the central mechanism of their development.
Collapse
Affiliation(s)
- Matthew Boyko
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Benjamin F Gruenbaum
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dmitry Frank
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Dmitry Natanel
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Shahar Negev
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Abed N Azab
- Department of Nursing, Recanati School for Community Health Professions, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Guy Barsky
- Department of Surgery B, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Boris Knyazer
- Department of Ophthalmology, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Ora Kofman
- Department of Psychology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Alexander Zlotnik
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| |
Collapse
|
25
|
Martynova E, Khaibullin T, Salafutdinov I, Markelova M, Laikov A, Lopukhov L, Liu R, Sahay K, Goyal M, Baranwal M, Rizvanov AA, Khaiboullina S. Seasonal Changes in Serum Metabolites in Multiple Sclerosis Relapse. Int J Mol Sci 2023; 24:3542. [PMID: 36834957 PMCID: PMC9959388 DOI: 10.3390/ijms24043542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Multiple sclerosis (MS) is a debilitating chronic disease of unknown etiology. There are limited treatment options due to an incomplete understanding of disease pathology. The disease is shown to have seasonal exacerbation of clinical symptoms. The mechanisms of such seasonal worsening of symptoms remains unknown. In this study, we applied targeted metabolomics analysis of serum samples using LC-MC/MC to determine seasonal changes in metabolites throughout the four seasons. We also analyzed seasonal serum cytokine alterations in patients with relapsed MS. For the first time, we can demonstrate seasonal changes in various metabolites in MS compared to the control. More metabolites were affected in MS in the fall season followed by spring, while summer MS was characterized by the smallest number of affected metabolites. Ceramides were activated in all seasons, suggesting their central role in the disease pathogenesis. Substantial changes in glucose metabolite levels were found in MS, indicating a potential shift to glycolysis. An increased serum level of quinolinic acid was demonstrated in winter MS. Histidine pathways were affected, suggesting their role in relapse of MS in the spring and fall. We also found that spring and fall seasons had a higher number of overlapping metabolites affected in MS. This could be explained by patients having a relapse of symptoms during these two seasons.
Collapse
Affiliation(s)
- Ekaterina Martynova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Timur Khaibullin
- Republican Clinical Neurological Center, Republic of Tatarstan, 420021 Kazan, Russia
| | - Ilnur Salafutdinov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
- Department of Medical Biology and Genetic, Kazan State Medical University, 420088 Kazan, Russia
| | - Maria Markelova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Alexander Laikov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Leonid Lopukhov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Rongzeng Liu
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471003, China
| | - Kritika Sahay
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Mehendi Goyal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Svetlana Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| |
Collapse
|
26
|
Application of Metabolic Reprogramming to Cancer Imaging and Diagnosis. Int J Mol Sci 2022; 23:ijms232415831. [PMID: 36555470 PMCID: PMC9782057 DOI: 10.3390/ijms232415831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Cellular metabolism governs the signaling that supports physiological mechanisms and homeostasis in an individual, including neuronal transmission, wound healing, and circadian clock manipulation. Various factors have been linked to abnormal metabolic reprogramming, including gene mutations, epigenetic modifications, altered protein epitopes, and their involvement in the development of disease, including cancer. The presence of multiple distinct hallmarks and the resulting cellular reprogramming process have gradually revealed that these metabolism-related molecules may be able to be used to track or prevent the progression of cancer. Consequently, translational medicines have been developed using metabolic substrates, precursors, and other products depending on their biochemical mechanism of action. It is important to note that these metabolic analogs can also be used for imaging and therapeutic purposes in addition to competing for metabolic functions. In particular, due to their isotopic labeling, these compounds may also be used to localize and visualize tumor cells after uptake. In this review, the current development status, applicability, and limitations of compounds targeting metabolic reprogramming are described, as well as the imaging platforms that are most suitable for each compound and the types of cancer to which they are most appropriate.
Collapse
|
27
|
Moss DY, McCann C, Kerr EM. Rerouting the drug response: Overcoming metabolic adaptation in KRAS-mutant cancers. Sci Signal 2022; 15:eabj3490. [PMID: 36256706 DOI: 10.1126/scisignal.abj3490] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mutations in guanosine triphosphatase KRAS are common in lung, colorectal, and pancreatic cancers. The constitutive activity of mutant KRAS and its downstream signaling pathways induces metabolic rewiring in tumor cells that can promote resistance to existing therapeutics. In this review, we discuss the metabolic pathways that are altered in response to treatment and those that can, in turn, alter treatment efficacy, as well as the role of metabolism in the tumor microenvironment (TME) in dictating the therapeutic response in KRAS-driven cancers. We highlight metabolic targets that may provide clinical opportunities to overcome therapeutic resistance and improve survival in patients with these aggressive cancers.
Collapse
Affiliation(s)
- Deborah Y Moss
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE Northern Ireland, UK
| | - Christopher McCann
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE Northern Ireland, UK
| | - Emma M Kerr
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE Northern Ireland, UK
| |
Collapse
|
28
|
Snyder MN, Henderson WM, Glinski DA, Purucker ST. Differentiating metabolomic responses of amphibians to multiple stressors. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 838:155666. [PMID: 35598671 PMCID: PMC9875051 DOI: 10.1016/j.scitotenv.2022.155666] [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: 08/02/2021] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
One of the biggest challenges in ecological risk assessment is determining the impact of multiple stressors on individual organisms and populations in real world scenarios. Frequently, data derived from laboratory studies of single stressors are used to estimate risk parameters and do not adequately address scenarios where other stressors exist. Emerging 'omic technologies, notably metabolomics, provide an opportunity to address the uncertainties surrounding ecological risk assessment of multiple stressors. The objective of this study was to use metabolomic profiling to investigate the effect of multiple stressors on amphibian metamorphs. We exposed post-metamorphosis (180 days) southern leopard frogs (Lithobates sphenocephala) to the insecticide carbaryl (480 μg/L), predation stress, and a combined pesticide and predation stress treatment. Corticosterone analysis revealed mild support for an induction in response to predation stress alone but strongly suggests that carbaryl exposure, alone or in combination with predation cues, can significantly elevate this known biomarker in amphibians. Metabolomics analysis accurately classed, based on relative nearness, carbaryl and predation induced changes in the hepatic metabolome and biochemical fluxes appear to be associated with a similar biological response. Support vector machine analysis with recursive feature elimination of the acquired metabolomic spectra demonstrated 85-96% classification accuracy among control and all treatment groups when using the top 75 ranked retention time bins. Biochemical fluxes observed in the groups exposed to carbaryl, predation, and the combined treatment include amino acids, sugar derivatives, and purine nucleotides. Ultimately, this methodology could be used to interpret short-term toxicity assays and the presence of environmental stressors to overall metabolomic effects in non-target organisms.
Collapse
Affiliation(s)
- Marcía N Snyder
- U.S. Environmental Protection Agency, ORD/CPHEA, Corvallis, OR 97333, USA.
| | | | - Donna A Glinski
- NRC Postdoctoral Research Fellow with the U.S. Environmental Protection Agency, Athens, GA 30605, USA.
| | - S Thomas Purucker
- U.S. Environmental Protection Agency, ORD/CCTE, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
29
|
Liu X, Zhang L, Zhang W. Metabolic reprogramming: A novel metabolic model for pulmonary hypertension. Front Cardiovasc Med 2022; 9:957524. [PMID: 36093148 PMCID: PMC9458918 DOI: 10.3389/fcvm.2022.957524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension, or PAH, is a condition that is characterized by pulmonary artery pressures above 20 mmHg (at rest). In the treatment of PAH, the pulmonary vascular system is regulated to ensure a diastolic and contraction balance; nevertheless, this treatment does not prevent or reverse pulmonary vascular remodeling and still causes pulmonary hypertension to progress. According to Warburg, the link between metabolism and proliferation in PAH is similar to that of cancer, with a common aerobic glycolytic phenotype. By activating HIF, aerobic glycolysis is enhanced and cell proliferation is triggered. Aside from glutamine metabolism, the Randle cycle is also present in PAH. Enhanced glutamine metabolism replenishes carbon intermediates used by glycolysis and provides energy to over-proliferating and anti-apoptotic pulmonary vascular cells. By activating the Randle cycle, aerobic oxidation is enhanced, ATP is increased, and myocardial injury is reduced. PAH is predisposed by epigenetic dysregulation of DNA methylation, histone acetylation, and microRNA. This article discusses the abnormal metabolism of PAH and how metabolic therapy can be used to combat remodeling.
Collapse
|
30
|
B 0 Correction for 3T Amide Proton Transfer (APT) MRI Using a Simplified Two-Pool Lorentzian Model of Symmetric Water and Asymmetric Solutes. Tomography 2022; 8:1974-1986. [PMID: 36006063 PMCID: PMC9412582 DOI: 10.3390/tomography8040165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/17/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
Amide proton transfer (APT)-weighted MRI is a promising molecular imaging technique that has been employed in clinic for detection and grading of brain tumors. MTRasym, the quantification method of APT, is easily influenced by B0 inhomogeneity and causes artifacts. Current model-free interpolation methods have enabled moderate B0 correction for middle offsets, but have performed poorly at limbic offsets. To address this shortcoming, we proposed a practical B0 correction approach that is suitable under time-limited sparse acquisition scenarios and for B1 ≥ 1 μT under 3T. In this study, this approach employed a simplified Lorentzian model containing only two pools of symmetric water and asymmetric solutes, to describe the Z-spectral shape with wide and ‘invisible’ CEST peaks. The B0 correction was then performed on the basis of the fitted two-pool Lorentzian lines, instead of using conventional model-free interpolation. The approach was firstly evaluated on densely sampled Z-spectra data by using the spline interpolation of all acquired 16 offsets as the gold standard. When only six offsets were available for B0 correction, our method outperformed conventional methods. In particular, the errors at limbic offsets were significantly reduced (n = 8, p < 0.01). Secondly, our method was assessed on the six-offset APT data of nine brain tumor patients. Our MTRasym (3.5 ppm), using the two-pool model, displayed a similar contrast to the vendor-provided B0-orrected MTRasym (3.5 ppm). While the vendor failed in correcting B0 at 4.3 and 2.7 ppm for a large portion of voxels, our method enabled well differentiation of B0 artifacts from tumors. In conclusion, the proposed approach could alleviate analysis errors caused by B0 inhomogeneity, which is useful for facilitating the comprehensive metabolic analysis of brain tumors.
Collapse
|
31
|
Špirková A, Kovaříková V, Šefčíková Z, Pisko J, Kšiňanová M, Koppel J, Fabian D, Čikoš Š. Glutamate can act as a signaling molecule in mouse preimplantation embryos. Biol Reprod 2022; 107:916-927. [PMID: 35746896 PMCID: PMC9562114 DOI: 10.1093/biolre/ioac126] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/31/2022] [Accepted: 06/20/2022] [Indexed: 11/12/2022] Open
Abstract
Free amino acids are present in the natural environment of the preimplantation embryo, and their availability can influence early embryo development. Glutamic acid is one of the amino acids with highest concentrations in female reproductive fluids, and we investigated whether glutamic acid/glutamate can affect preimplantation embryo development by acting through cell membrane receptors. Using RT-PCR, we detected 15 ionotropic glutamate receptor transcripts and 8 metabotropic glutamate receptor transcripts in mouse ovulated oocytes and/or in vivo developed blastocysts. Using immunohistochemistry, we detected expression of two AMPA receptor subunits, three kainate receptor subunits and member 5 metabotropic glutamate receptor protein in blastocysts. Extracellular concentrations of glutamic acid starting at 5 mM impaired mouse blastocyst development, and this fact may be of great practical importance since glutamic acid and its salts (mainly monosodium glutamate) are widely used as food additives. Experiments with glutamate receptor agonists (in combination with gene expression analysis) revealed that specific AMPA receptors (formed from GRIA3 and/or GRIA4 subunits), kainate receptors (formed from GRIK 3 and GRIK 4 or GRIK 5 subunits) and GRM5 glutamate receptor were involved in this effect. The glutamic acid-induced effects were prevented or reduced by pre-treatment of blastocysts with AMPA, kainate and GRM5 receptor antagonists, further confirming the involvement of these receptor types. Our results show that glutamic acid can act as a signaling molecule in preimplantation embryos, exerting its effects through activation of cell membrane receptors.
Collapse
Affiliation(s)
- Alexandra Špirková
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Veronika Kovaříková
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Zuzana Šefčíková
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Jozef Pisko
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Martina Kšiňanová
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| | - Juraj Koppel
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Dušan Fabian
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Štefan Čikoš
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| |
Collapse
|
32
|
Alsoud LO, Soares NC, Al-Hroub HM, Mousa M, Kasabri V, Bulatova N, Suyagh M, Alzoubi KH, El-Huneidi W, Abu-Irmaileh B, Bustanji Y, Semreen MH. Identification of Insulin Resistance Biomarkers in Metabolic Syndrome Detected by UHPLC-ESI-QTOF-MS. Metabolites 2022; 12:metabo12060508. [PMID: 35736441 PMCID: PMC9227428 DOI: 10.3390/metabo12060508] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/28/2022] [Accepted: 05/29/2022] [Indexed: 02/04/2023] Open
Abstract
Metabolic syndrome (MetS) is a disorder characterized by a group of factors that can increase the risk of chronic diseases, including cardiovascular diseases and type 2 diabetes mellitus (T2D). Metabolomics has provided new insight into disease diagnosis and biomarker identification. This cross-sectional investigation used an untargeted metabolomics-based technique to uncover metabolomic alterations and their relationship to pathways in normoglycemic and prediabetic MetS participants to improve disease diagnosis. Plasma samples were collected from drug-naive prediabetic MetS patients (n = 26), normoglycemic MetS patients (n = 30), and healthy (normoglycemic lean) subjects (n = 30) who met the inclusion criteria for the study. The plasma samples were analyzed using highly sensitive ultra-high-performance liquid chromatography electrospray ionization quadrupole time-of-flight mass spectrometry (UHPLC-ESI-QTOF-MS). One-way ANOVA analysis revealed that 59 metabolites differed significantly among the three groups (p < 0.05). Glutamine, 5-hydroxy-L-tryptophan, L-sorbose, and hippurate were highly associated with MetS. However, 9-methyluric acid, sphinganine, and threonic acid were highly associated with prediabetes/MetS. Metabolic pathway analysis showed that arginine biosynthesis and glutathione metabolism were associated with MetS/prediabetes, while phenylalanine, D-glutamine and D-glutamate, and lysine degradation were highly impacted in MetS. The current study sheds light on the potential diagnostic value of some metabolites in metabolic syndrome and the role of their alteration on some of the metabolic pathways. More studies are needed in larger cohorts in order to verify the implication of the above metabolites on MetS and their diagnostic value.
Collapse
Affiliation(s)
- Leen Oyoun Alsoud
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (L.O.A.); (N.C.S.); (K.H.A.)
| | - Nelson C. Soares
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (L.O.A.); (N.C.S.); (K.H.A.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (H.M.A.-H.); (W.E.-H.)
| | - Hamza M. Al-Hroub
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (H.M.A.-H.); (W.E.-H.)
| | - Muath Mousa
- Research Institute of Science and Engineering, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates;
| | - Violet Kasabri
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (V.K.); (N.B.); (M.S.)
| | - Nailya Bulatova
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (V.K.); (N.B.); (M.S.)
| | - Maysa Suyagh
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (V.K.); (N.B.); (M.S.)
| | - Karem H. Alzoubi
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (L.O.A.); (N.C.S.); (K.H.A.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (H.M.A.-H.); (W.E.-H.)
| | - Waseem El-Huneidi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (H.M.A.-H.); (W.E.-H.)
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Bashaer Abu-Irmaileh
- Hamdi Mango Center for Scientific Research, The University of Jordan, Amman 11942, Jordan;
| | - Yasser Bustanji
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (H.M.A.-H.); (W.E.-H.)
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (V.K.); (N.B.); (M.S.)
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Correspondence: (Y.B.); (M.H.S.)
| | - Mohammad H. Semreen
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (L.O.A.); (N.C.S.); (K.H.A.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (H.M.A.-H.); (W.E.-H.)
- Correspondence: (Y.B.); (M.H.S.)
| |
Collapse
|
33
|
Repetitive Transcranial Magnetic Stimulation-Associated Changes in Neocortical Metabolites in Major Depression: A Systematic Review. Neuroimage Clin 2022; 35:103049. [PMID: 35738081 PMCID: PMC9233277 DOI: 10.1016/j.nicl.2022.103049] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/01/2022] [Accepted: 05/12/2022] [Indexed: 11/23/2022]
Abstract
We reviewed 12 studies that measured metabolites pre and post rTMS in MDD. Frontal lobe Glu, Gln, NAA, and GABA increased after rTMS. Increases in metabolites were often associated with MDD symptom improvement. We propose novel intracellular mechanisms by which metabolites are altered by rTMS.
Introduction Repetitive Transcranial magnetic stimulation (rTMS) is an FDA approved treatment for major depressive disorder (MDD). However, neural mechanisms contributing to rTMS effects on depressive symptoms, cognition, and behavior are unclear. Proton magnetic resonance spectroscopy (MRS), a noninvasive neuroimaging technique measuring concentrations of biochemical compounds within the brain in vivo, may provide mechanistic insights. Methods This systematic review summarized published MRS findings from rTMS treatment trials to address potential neurometabolic mechanisms of its antidepressant action. Using PubMed, Google Scholar, Web of Science, and JSTOR, we identified twelve empirical studies that evaluated changes in MRS metabolites in a within-subjects, pre- vs. post-rTMS treatment design in patients with MDD. Results rTMS protocols ranged from four days to eight weeks duration, were applied at high frequency to the left dorsolateral prefrontal cortex (DLPFC) in most studies, and were conducted in patients aged 13-to-70. Most studies utilized MRS point resolved spectroscopy acquisitions at 3 Tesla in the bilateral anterior cingulate cortex and DLPFC. Symptom improvements were correlated with rTMS-related increases in the concentration of glutamatergic compounds (glutamate, Glu, and glutamine, Gln), GABA, and N-acetylated compounds (NAA), with some results trend-level. Conclusions This is the first in-depth systematic review of metabolic effects of rTMS in individuals with MDD. The extant literature suggests rTMS stimulation does not produce changes in neurometabolites independent of clinical response; increases in frontal lobe glutamatergic compounds, N-acetylated compounds and GABA following high frequency left DLPFC rTMS therapy were generally associated with clinical improvement. Glu, Gln, GABA, and NAA may mediate rTMS treatment effects on MDD symptomatology through intracellular mechanisms.
Collapse
|
34
|
Comprehensive Metabolic Profiling of MYC-Amplified Medulloblastoma Tumors Reveals Key Dependencies on Amino Acid, Tricarboxylic Acid and Hexosamine Pathways. Cancers (Basel) 2022; 14:cancers14051311. [PMID: 35267619 PMCID: PMC8909278 DOI: 10.3390/cancers14051311] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/21/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary The oncogene MYC alters cellular metabolism. Medulloblastoma is the most common malignant pediatric brain tumor. MYC-amplified medulloblastoma has a poor prognosis, and the metabolism of MYC-amplified medulloblastoma is poorly understood. We performed comprehensive metabolic profiling of MYC-amplified medulloblastoma and found increased reliance on potentially targetable pathways. We also found that the metabolism of MYC-amplified cell lines differed from orthotopic brain tumors in vitro and in flank tumors, suggesting that analyses conducted in vitro or in flank tumors may miss key vulnerabilities. Abstract Reprograming of cellular metabolism is a hallmark of cancer. Altering metabolism allows cancer cells to overcome unfavorable microenvironment conditions and to proliferate and invade. Medulloblastoma is the most common malignant brain tumor of children. Genomic amplification of MYC defines a subset of poor-prognosis medulloblastoma. We performed comprehensive metabolic studies of human MYC-amplified medulloblastoma by comparing the metabolic profiles of tumor cells in three different conditions—in vitro, in flank xenografts and in orthotopic xenografts in the cerebellum. Principal component analysis showed that the metabolic profiles of brain and flank high-MYC medulloblastoma tumors clustered closely together and separated away from normal brain and in vitro MYC-amplified cells. Compared to normal brain, MYC-amplified medulloblastoma orthotopic xenograft tumors showed upregulation of the TCA cycle as well as the synthesis of nucleotides, hexosamines, amino acids and glutathione. There was significantly higher glucose uptake and usage in orthotopic xenograft tumors compared to flank xenograft tumors and cells in culture. In orthotopic tumors, glucose was the main carbon source for the de novo synthesis of glutamate, glutamine and glutathione through the TCA cycle. In vivo, the glutaminase II pathway was the main pathway utilizing glutamine. Glutathione was the most abundant upregulated metabolite in orthotopic tumors compared to normal brain. Glutamine-derived glutathione was synthesized through the glutamine transaminase K (GTK) enzyme in vivo. In conclusion, high MYC medulloblastoma cells have different metabolic profiles in vitro compared to in vivo, and key vulnerabilities may be missed by not performing in vivo metabolic analyses.
Collapse
|
35
|
Veshkini A, M Hammon H, Vogel L, Delosière M, Viala D, Dèjean S, Tröscher A, Ceciliani F, Sauerwein H, Bonnet M. Liver proteome profiling in dairy cows during the transition from gestation to lactation: Effects of supplementation with essential fatty acids and conjugated linoleic acids as explored by PLS-DA. J Proteomics 2022; 252:104436. [PMID: 34839038 DOI: 10.1016/j.jprot.2021.104436] [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: 06/14/2021] [Revised: 10/11/2021] [Accepted: 11/09/2021] [Indexed: 01/08/2023]
Abstract
This study aimed at investigating the synergistic effects of essential fatty acids (EFA) and conjugated linoleic acids (CLA) on the liver proteome profile of dairy cows during the transition to lactation. 16 Holstein cows were infused from 9 wk. antepartum to 9 wk. postpartum into the abomasum with either coconut oil (CTRL) or a mixture of EFA (linseed + safflower oil) and CLA (EFA + CLA). Label-free quantitative proteomics was performed in liver tissue biopsied at days -21, +1, +28, and + 63 relative to calving. Differentially abundant proteins (DAP) between treatment groups were identified at the intersection between a multivariate and a univariate analysis. In total, 1680 proteins were identified at each time point, of which between groups DAP were assigned to the metabolism of xenobiotics by cytochrome P450, drug metabolism - cytochrome P450, steroid hormone biosynthesis, glycolysis/gluconeogenesis, and glutathione metabolism. Cytochrome P450, as a central hub, enriched with specific CYP enzymes comprising: CYP51A1 (d - 21), CYP1A1 & CYP4F2 (d + 28), and CYP4V2 (d + 63). Collectively, supplementation of EFA + CLA in transition cows impacted hepatic lipid metabolism and enriched several common biological pathways at all time points that were mainly related to ω-oxidation of fatty acids through the Cytochrome p450 pathway. SIGNIFICANCE: In three aspects this manuscript is notable. First, this is among the first longitudinal proteomics studies in nutrition of dairy cows. The selected time points are critical periods around parturition with profound endocrine and metabolic adaptations. Second, our findings provided novel information on key drivers of biologically relevant pathways suggested according to previously reported performance, zootechnical, and metabolism data (already published elsewhere). Third, our results revealed the role of cytochrome P450 that is hardly investigated, and of ω-oxidation pathways in the metabolism of fatty acids with the involvement of specific enzymes.
Collapse
Affiliation(s)
- Arash Veshkini
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn, Germany; Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; INRAE, Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France; Department of Veterinary Medicine, Università degli Studi di Milano, Lodi, Italy
| | - Harald M Hammon
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany.
| | - Laura Vogel
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Mylène Delosière
- INRAE, Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France
| | - Didier Viala
- INRAE, Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France
| | - Sèbastien Dèjean
- Institut de Mathématiques de Toulouse, UMR5219, Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | | | - Fabrizio Ceciliani
- Department of Veterinary Medicine, Università degli Studi di Milano, Lodi, Italy
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn, Germany
| | - Muriel Bonnet
- INRAE, Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France.
| |
Collapse
|
36
|
Duft RG, Castro A, Bonfante ILP, Lopes WA, da Silva LR, Chacon-Mikahil MPT, Leite N, Cavaglieri CR. Serum metabolites associated with increased insulin resistance and low cardiorespiratory fitness in overweight adolescents. Nutr Metab Cardiovasc Dis 2022; 32:269-278. [PMID: 34906412 DOI: 10.1016/j.numecd.2021.09.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Obesity affects metabolism, increasing the risk of developing non-communicable diseases in adolescence, due to excess adipose tissue and low cardiorespiratory fitness (CRF). The metabolomics approach allows the elucidation of metabolites, which may have the concentrations altered by several factors, such as body composition (BC). We aimed to analyze the metabolomic profile of normal-weight and overweight adolescents and associate the metabolites with clinical markers related to BC, insulin resistance (IR), and CRF. METHODS AND RESULTS The sample was composed of 57 adolescents and divided into two groups: the normal-weight group (NWG, n = 24) and the overweight group (OWG, n = 33). They underwent blood collection and anthropometric, BC, and CRF assessment. Blood serum samples were analyzed by 1H-NMR spectroscopy (600 MHz). The OWG presented higher values of body weight (BW), body mass index (BMI), waist circumference (WC), fasting glucose, insulin, IR, cholesterol, and percentage of fat mass (%FM) and lower levels of peak oxygen consumption (VO2 peak) compared with the NWG. The OWG presented lower concentrations of 3-hydroxyisovalerate, glutamate, and methionine as well as higher concentrations of aspartate, asparagine, creatine, glycerol, myo-inositol, proline, pyruvate, tyrosine, and valerate compared with the NWG. The concentrations of glutamate, myo-inositol, creatine, methionine, and valerate correlated with %FM; pyruvate and valerate positively correlated with IR; and glutamate, tyrosine, and valerate negatively correlated with CRF. CONCLUSION Changes in the BC lead to changes in the metabolomic profile of adolescents, and the altered metabolites are associated with increased IR and low CRF. These results indicate new targets for health monitoring and prevention of cardiovascular and metabolic diseases in adolescents.
Collapse
Affiliation(s)
- Renata G Duft
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, Campinas, São Paulo, Brazil.
| | - Alex Castro
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, Campinas, São Paulo, Brazil
| | - Ivan L P Bonfante
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, Campinas, São Paulo, Brazil; Federal Institute of Education, Science and Technology of São Paulo, Hortolandia Campus, Hortolandia, Brazil
| | - Wendell A Lopes
- Department of Physical Education, State University of Maringa, Maringa, Brazil
| | - Larissa R da Silva
- Department of Physical Education, University of Parana, Curitiba, Brazil
| | - Mara P T Chacon-Mikahil
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, Campinas, São Paulo, Brazil
| | - Neiva Leite
- Department of Physical Education, University of Parana, Curitiba, Brazil
| | - Cláudia R Cavaglieri
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
37
|
Singh PP, Srivastava D, Shukla S, Varsha. Rhizophagus proliferus genome sequence reiterates conservation of genetic traits in AM fungi, but predicts higher saprotrophic activity. Arch Microbiol 2021; 204:105. [DOI: 10.1007/s00203-021-02651-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/24/2022]
|
38
|
Hu JP, Zheng TT, Zeng BF, Wu ML, Shi R, Zhang Y, Chen LJ, Cheng WJ, Liang P. Effects of Lactobacillus plantarum FZU3013-Fermented Laminaria japonica on Lipid Metabolism and Gut Microbiota in Hyperlipidaemic Rats. Front Nutr 2021; 8:786571. [PMID: 34938762 PMCID: PMC8685254 DOI: 10.3389/fnut.2021.786571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
In this study, we explored the effect of Lactobacillus plantarum FZU3013-fermented Laminaria japonica (LPLJ) supplementation to prevent hyperlipidaemia in rats fed with a high-fat diet (HFD). The results indicate that LPLJ supplementation improved serum and hepatic biochemical indicators (p < 0.05), elevated short-chain fatty acid levels, reduced HFD-induced accumulation of lipid droplets in the liver, modulated the relative abundance of some microbial phylotypes, and reduced hyperlipidaemia in HFD-fed rats by adjusting the aminoacyl-tRNA, phenylalanine, tyrosine, and tryptophan biosynthetic pathways, as well as the phenylalanine, D-glutamine and D-glutamate, and glutathione metabolic pathways. Additionally, hepatic mRNA levels of the genes involved in lipid metabolism and bile acid homeostasis were significantly reduced by LPLJ intervention (p < 0.05). These results suggest that LPLJ has a positive effect on modulating lipid metabolism and has the potential to be a functional food that can help prevent hyperlipidaemia.
Collapse
Affiliation(s)
- Jin-Peng Hu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ting-Ting Zheng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Bin-Fen Zeng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Man-Ling Wu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Rui Shi
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ye Zhang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Li-Jiao Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Wen-Jian Cheng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Peng Liang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
39
|
Ordoñez JFF, Galindez GGST, Gulay KT, Ravago-Gotanco R. Transcriptome analysis of growth variation in early juvenile stage sandfish Holothuria scabra. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2021; 40:100904. [PMID: 34488170 DOI: 10.1016/j.cbd.2021.100904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/28/2021] [Accepted: 08/14/2021] [Indexed: 06/13/2023]
Abstract
The sandfish Holothuria scabra is a high-value tropical sea cucumber species representing a major mariculture prospect across the Indo-Pacific. Advancements in culture technology, rearing, and processing present options for augmenting capture production, stock restoration, and sustainable livelihood activities from hatchery-produced sandfish. Further improvements in mariculture production may be gained from the application of genomic technologies to improve performance traits such as growth. In this study, we performed de novo transcriptome assembly and characterization of fast- and slow-growing juvenile H. scabra from three Philippine populations. Analyses revealed 66 unigenes that were consistently differentially regulated in fast-growing sandfish and found to be associated with immune response and metabolism. Further, we identified microsatellite and single nucleotide polymorphism markers potentially associated with fast growth. These findings provide insight on potential genomic determinants underlying growth regulation in early juvenile sandfish which will be useful for further functional studies.
Collapse
Affiliation(s)
- June Feliciano F Ordoñez
- The Marine Science Institute, University of the Philippines Diliman, Velasquez St., Diliman, 1100 Quezon City, Philippines.
| | - Gihanna Gaye S T Galindez
- The Marine Science Institute, University of the Philippines Diliman, Velasquez St., Diliman, 1100 Quezon City, Philippines; Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Hannover Medical School, Germany.
| | - Karina Therese Gulay
- The Marine Science Institute, University of the Philippines Diliman, Velasquez St., Diliman, 1100 Quezon City, Philippines.
| | - Rachel Ravago-Gotanco
- The Marine Science Institute, University of the Philippines Diliman, Velasquez St., Diliman, 1100 Quezon City, Philippines.
| |
Collapse
|
40
|
Mondal M, Basak S, Choudhury S, Ghosh NN, Roy MN. Investigation of molecular interactions insight into some biologically active amino acids and aqueous solutions of an anti-malarial drug by physicochemical and theoretical approach. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116933] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
41
|
Moroz LL, Nikitin MA, Poličar PG, Kohn AB, Romanova DY. Evolution of glutamatergic signaling and synapses. Neuropharmacology 2021; 199:108740. [PMID: 34343611 PMCID: PMC9233959 DOI: 10.1016/j.neuropharm.2021.108740] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022]
Abstract
Glutamate (Glu) is the primary excitatory transmitter in the mammalian brain. But, we know little about the evolutionary history of this adaptation, including the selection of l-glutamate as a signaling molecule in the first place. Here, we used comparative metabolomics and genomic data to reconstruct the genealogy of glutamatergic signaling. The origin of Glu-mediated communications might be traced to primordial nitrogen and carbon metabolic pathways. The versatile chemistry of L-Glu placed this molecule at the crossroad of cellular biochemistry as one of the most abundant metabolites. From there, innovations multiplied. Many stress factors or injuries could increase extracellular glutamate concentration, which led to the development of modular molecular systems for its rapid sensing in bacteria and archaea. More than 20 evolutionarily distinct families of ionotropic glutamate receptors (iGluRs) have been identified in eukaryotes. The domain compositions of iGluRs correlate with the origins of multicellularity in eukaryotes. Although L-Glu was recruited as a neuro-muscular transmitter in the early-branching metazoans, it was predominantly a non-neuronal messenger, with a possibility that glutamatergic synapses evolved more than once. Furthermore, the molecular secretory complexity of glutamatergic synapses in invertebrates (e.g., Aplysia) can exceed their vertebrate counterparts. Comparative genomics also revealed 15+ subfamilies of iGluRs across Metazoa. However, most of this ancestral diversity had been lost in the vertebrate lineage, preserving AMPA, Kainate, Delta, and NMDA receptors. The widespread expansion of glutamate synapses in the cortical areas might be associated with the enhanced metabolic demands of the complex brain and compartmentalization of Glu signaling within modular neuronal ensembles.
Collapse
Affiliation(s)
- Leonid L Moroz
- Whitney Laboratory for Marine Biosciences, University of Florida, St. Augustine, FL, 32080, USA; Departments of Neuroscience and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| | - Mikhail A Nikitin
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119991, Russia; Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, 127994, Russia
| | - Pavlin G Poličar
- Whitney Laboratory for Marine Biosciences, University of Florida, St. Augustine, FL, 32080, USA; Faculty of Computer and Information Science, University of Ljubljana, SI-1000, Ljubljana, Slovenia
| | - Andrea B Kohn
- Whitney Laboratory for Marine Biosciences, University of Florida, St. Augustine, FL, 32080, USA
| | - Daria Y Romanova
- Cellular Neurobiology of Learning Lab, Institute of Higher Nervous Activity and Neurophysiology, Moscow, 117485, Russia.
| |
Collapse
|
42
|
Glutamine Homeostasis and Its Role in the Adaptive Strategies of the Blind Mole Rat, Spalax. Metabolites 2021; 11:metabo11110755. [PMID: 34822413 PMCID: PMC8620300 DOI: 10.3390/metabo11110755] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 10/26/2021] [Accepted: 10/30/2021] [Indexed: 12/20/2022] Open
Abstract
Oxidative metabolism is fine-tuned machinery that combines two tightly coupled fluxes of glucose and glutamine-derived carbons. Hypoxia interrupts the coordination between the metabolism of these two nutrients and leads to a decrease of the system efficacy and may eventually cause cell death. The subterranean blind mole rat, Spalax, is an underexplored, underground, hypoxia-tolerant mammalian group which spends its life under sharply fluctuating oxygen levels. Primary Spalax cells are an exceptional model to study the metabolic strategies that have evolved in mammals inhabiting low-oxygen niches. In this study we explored the metabolic frame of glutamine (Gln) homeostasis in Spalax skin cells under normoxic and hypoxic conditions and their impacts on the metabolism of rat cells. Targeted metabolomics employing liquid chromatography and mass spectrometry (LC-MS) was used to track the fate of heavy glutamine carbons (13C5 Gln) after 24 h under normoxia or hypoxia (1% O2). Our results indicated that large amounts of glutamine-originated carbons were detected as proline (Pro) and hydroxyproline (HPro) in normoxic Spalax cells with a further increase under hypoxia, suggesting a strategy for reduced Gln carbons storage in proteins. The intensity of the flux and the presence of HPro suggests collagen as a candidate protein that is most abundant in animals, and as the primary source of HPro. An increased conversion of αKG to 2 HG that was indicated in hypoxic Spalax cells prevents the degradation of hypoxia-inducible factor 1α (HIF-1α) and, consequently, maintains cytosolic and mitochondrial carbons fluxes that were uncoupled via inhibition of the pyruvate dehydrogenase complex. A strong antioxidant defense in Spalax cells can be attributed, at least in part, to the massive usage of glutamine-derived glutamate for glutathione (GSH) production. The present study uncovers additional strategies that have evolved in this unique mammal to support its hypoxia tolerance, and probably contribute to its cancer resistance, longevity, and healthy aging.
Collapse
|
43
|
Miner MWG, Liljenbäck H, Virta J, Helin S, Eskola O, Elo P, Teuho J, Seppälä K, Oikonen V, Yang G, Kindler-Röhrborn A, Minn H, Li XG, Roivainen A. Comparison of: (2 S,4 R)-4-[ 18F]Fluoroglutamine, [ 11C]Methionine, and 2-Deoxy-2-[ 18F]Fluoro- D-Glucose and Two Small-Animal PET/CT Systems Imaging Rat Gliomas. Front Oncol 2021; 11:730358. [PMID: 34692505 PMCID: PMC8530378 DOI: 10.3389/fonc.2021.730358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/14/2021] [Indexed: 12/04/2022] Open
Abstract
Purpose The three positron emission tomography (PET) imaging compounds: (2S,4R)-4-[18F]Fluoroglutamine ([18F]FGln), L-[methyl-11C]Methionine ([11C]Met), and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) were investigated to contrast their ability to image orthotopic BT4C gliomas in BDIX rats. Two separate small animal imaging systems were compared for their tumor detection potential. Dynamic acquisition of [18F]FGln was evaluated with multiple pharmacokinetic models for future quantitative comparison. Procedures Up to four imaging studies were performed on each orthotopically grafted BT4C glioma-bearing BDIX rat subject (n = 16) on four consecutive days. First, a DOTAREM® contrast enhanced MRI followed by attenuation correction CT and dynamic PET imaging with each radiopharmaceutical (20 min [11C]Met, 60 min [18F]FDG, and 60 min [18F]FGln with either the Molecubes PET/CT (n = 5) or Inveon PET/CT cameras (n = 11). Ex vivo brain autoradiography was completed for each radiopharmaceutical and [18F]FGln pharmacokinetics were studied by injecting 40 MBq into healthy BDIX rats (n = 10) and collecting blood samples between 5 and 60 min. Erythrocyte uptake, plasma protein binding and plasma parent-fraction were combined to estimate the total blood bioavailability of [18F]FGln over time. The corrected PET-image blood data was then applied to multiple pharmacokinetic models. Results Average BT4C tumor-to-healthy brain tissue uptake ratios (TBR) for PET images reached maxima of: [18F]FGln TBR: 1.99 ± 0.19 (n = 13), [18F]FDG TBR: 1.41 ± 0.11 (n = 6), and [11C]Met TBR: 1.08 ± 0.08, (n = 12) for the dynamic PET images. Pharmacokinetic modeling in dynamic [18F]FGln studies suggested both reversible and irreversible uptake play a similar role. Imaging with Inveon and Molecubes yielded similar end-result ratios with insignificant differences (p > 0.25). Conclusions In orthotopic BT4C gliomas, [18F]FGln may offer improved imaging versus [11C]Met and [18F]FDG. No significant difference in normalized end-result data was found between the Inveon and Molecubes camera systems. Kinetic modelling of [18F]FGln uptake suggests that both reversible and irreversible uptake play an important role in BDIX rat pharmacokinetics.
Collapse
Affiliation(s)
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Jenni Virta
- Turku PET Centre, University of Turku, Turku, Finland
| | - Semi Helin
- Turku PET Centre, University of Turku, Turku, Finland
| | - Olli Eskola
- Turku PET Centre, University of Turku, Turku, Finland
| | - Petri Elo
- Turku PET Centre, University of Turku, Turku, Finland
| | - Jarmo Teuho
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Kerttu Seppälä
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Vesa Oikonen
- Turku PET Centre, University of Turku, Turku, Finland
| | - Guangli Yang
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Andrea Kindler-Röhrborn
- Institute of Pathology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Heikki Minn
- Turku PET Centre, University of Turku, Turku, Finland.,Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
| | - Xiang-Guo Li
- Turku PET Centre, University of Turku, Turku, Finland.,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland.,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| |
Collapse
|
44
|
Nassan FL, Kelly RS, Koutrakis P, Vokonas PS, Lasky-Su JA, Schwartz JD. Metabolomic signatures of the short-term exposure to air pollution and temperature. ENVIRONMENTAL RESEARCH 2021; 201:111553. [PMID: 34171372 PMCID: PMC8478827 DOI: 10.1016/j.envres.2021.111553] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 06/07/2021] [Accepted: 06/16/2021] [Indexed: 05/29/2023]
Abstract
BACKGROUND Short-term exposures to air pollution and temperature have been reported to be associated with inflammation and oxidative stress. However, mechanistic understanding of the affected metabolic pathways is still lacking and literature on the short-term exposure of air-pollution on the metabolome is limited. OBJECTIVES We aimed to determine changes in the plasma metabolome and associated metabolic pathways related to short-term exposure to outdoor air pollution and temperature. METHODS We performed mass-spectrometry based untargeted metabolomic profiling of plasma samples from a large and well-characterized cohort of men (Normative Aging Study) to identify metabolic pathways associated with short-term exposure to PM2.5, NO2, O3, and temperature (one, seven-, and thirty-day average of address-specific predicted estimates). We used multivariable linear mixed-effect regression and independent component analysis (ICA) while simultaneously adjusting for all exposures and correcting for multiple testing. RESULTS Overall, 456 white men provided 648 blood samples, in which 1158 metabolites were quantified, between 2000 and 2016. Average age and body mass index were 75.0 years and 27.7 kg/m2, respectively. Only 3% were current smokers. In the adjusted models, NO2, and temperature showed statistically significant associations with several metabolites (19 metabolites for NO2 and 5 metabolites for temperature). We identified six metabolic pathways (sphingolipid, butanoate, pyrimidine, glycolysis/gluconeogenesis, propanoate, and pyruvate metabolisms) perturbed with short-term exposure to air pollution and temperature. These pathways were involved in inflammation and oxidative stress, immunity, and nucleic acid damage and repair. CONCLUSIONS This is the first study to report an untargeted metabolomic signature of temperature exposure, the largest to report an untargeted metabolomic signature of air pollution, and the first to use ICA. We identified several significant plasma metabolites and metabolic pathways associated with short-term exposure to air pollution and temperature; using an untargeted approach. Those pathways were involved in inflammation and oxidative stress, immunity, and nucleic acid damage and repair. These results need to be confirmed by future research.
Collapse
Affiliation(s)
- Feiby L Nassan
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA.
| | - Rachel S Kelly
- Channing Division of Network Medicine; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Pantel S Vokonas
- VA Normative Aging Study, VA Boston Healthcare System, School of Medicine and School of Public Health, Boston University, Boston MA, 02215, USA
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Joel D Schwartz
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA; Channing Division of Network Medicine; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02129, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| |
Collapse
|
45
|
Shytaj IL, Procopio FA, Tarek M, Carlon‐Andres I, Tang H, Goldman AR, Munshi M, Kumar Pal V, Forcato M, Sreeram S, Leskov K, Ye F, Lucic B, Cruz N, Ndhlovu LC, Bicciato S, Padilla‐Parra S, Diaz RS, Singh A, Lusic M, Karn J, Alvarez‐Carbonell D, Savarino A. Glycolysis downregulation is a hallmark of HIV-1 latency and sensitizes infected cells to oxidative stress. EMBO Mol Med 2021; 13:e13901. [PMID: 34289240 PMCID: PMC8350904 DOI: 10.15252/emmm.202013901] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
HIV-1 infects lymphoid and myeloid cells, which can harbor a latent proviral reservoir responsible for maintaining lifelong infection. Glycolytic metabolism has been identified as a determinant of susceptibility to HIV-1 infection, but its role in the development and maintenance of HIV-1 latency has not been elucidated. By combining transcriptomic, proteomic, and metabolomic analyses, we here show that transition to latent HIV-1 infection downregulates glycolysis, while viral reactivation by conventional stimuli reverts this effect. Decreased glycolytic output in latently infected cells is associated with downregulation of NAD+ /NADH. Consequently, infected cells rely on the parallel pentose phosphate pathway and its main product, NADPH, fueling antioxidant pathways maintaining HIV-1 latency. Of note, blocking NADPH downstream effectors, thioredoxin and glutathione, favors HIV-1 reactivation from latency in lymphoid and myeloid cellular models. This provides a "shock and kill effect" decreasing proviral DNA in cells from people living with HIV/AIDS. Overall, our data show that downmodulation of glycolysis is a metabolic signature of HIV-1 latency that can be exploited to target latently infected cells with eradication strategies.
Collapse
Affiliation(s)
- Iart Luca Shytaj
- Department of Infectious DiseasesItalian Institute of HealthRomeItaly
- Department of Infectious DiseasesHeidelberg University HospitalHeidelbergGermany
- Infectious Diseases DepartmentFederal University of São PauloSão PauloBrazil
| | - Francesco Andrea Procopio
- Service of Immunology and AllergyLausanne University HospitalUniversity of LausanneLausanneSwitzerland
| | - Mohammad Tarek
- Bioinformatics DepartmentArmed Forces College of Medicine (AFCM)CairoEgypt
| | - Irene Carlon‐Andres
- Division of Structural BiologyWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
- Department of Infectious DiseasesFaculty of Life Sciences & MedicineKing’s College LondonLondonUK
- Randall Division of Cell and Molecular BiophysicsKing’s College LondonLondonUK
| | | | | | | | | | - Mattia Forcato
- Department of Life SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Sheetal Sreeram
- Department of Molecular Biology and MicrobiologyCase Western Reserve UniversityClevelandOHUSA
| | - Konstantin Leskov
- Department of Molecular Biology and MicrobiologyCase Western Reserve UniversityClevelandOHUSA
| | - Fengchun Ye
- Department of Molecular Biology and MicrobiologyCase Western Reserve UniversityClevelandOHUSA
| | - Bojana Lucic
- Department of Infectious DiseasesHeidelberg University HospitalHeidelbergGermany
- German Center for Infection ResearchHeidelbergGermany
| | - Nicolly Cruz
- Infectious Diseases DepartmentFederal University of São PauloSão PauloBrazil
| | - Lishomwa C Ndhlovu
- Division of Infectious DiseasesDepartment of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Silvio Bicciato
- Department of Life SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Sergi Padilla‐Parra
- Division of Structural BiologyWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
- Department of Infectious DiseasesFaculty of Life Sciences & MedicineKing’s College LondonLondonUK
- Randall Division of Cell and Molecular BiophysicsKing’s College LondonLondonUK
| | - Ricardo Sobhie Diaz
- Infectious Diseases DepartmentFederal University of São PauloSão PauloBrazil
| | - Amit Singh
- Indian Institute of ScienceBangaloreIndia
| | - Marina Lusic
- Department of Infectious DiseasesHeidelberg University HospitalHeidelbergGermany
- German Center for Infection ResearchHeidelbergGermany
| | - Jonathan Karn
- Department of Molecular Biology and MicrobiologyCase Western Reserve UniversityClevelandOHUSA
| | - David Alvarez‐Carbonell
- Department of Molecular Biology and MicrobiologyCase Western Reserve UniversityClevelandOHUSA
| | - Andrea Savarino
- Department of Infectious DiseasesItalian Institute of HealthRomeItaly
| |
Collapse
|
46
|
Nassan FL, Wang C, Kelly RS, Lasky-Su JA, Vokonas PS, Koutrakis P, Schwartz JD. Ambient PM 2.5 species and ultrafine particle exposure and their differential metabolomic signatures. ENVIRONMENT INTERNATIONAL 2021; 151:106447. [PMID: 33639346 PMCID: PMC7994935 DOI: 10.1016/j.envint.2021.106447] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/03/2021] [Indexed: 05/09/2023]
Abstract
BACKGROUND The metabolomic signatures of short- and long-term exposure to PM2.5 have been reported and linked to inflammation and oxidative stress. However, little is known about the relative contribution of the specific PM2.5 species (hence sources) that drive these metabolomic signatures. OBJECTIVES We aimed to determine the relative contribution of the different species of PM2.5 exposure to the perturbed metabolic pathways related to changes in the plasma metabolome. METHODS We performed mass-spectrometry based metabolomic profiling of plasma samples among men from the Normative Aging Study to identify metabolic pathways associated with PM2.5 species. The exposure windows included short-term (one, seven-, and thirty-day moving average) and long-term (one year moving average). We used linear mixed-effect regression with subject-specific intercepts while simultaneously adjusting for PM2.5, NO2, O3, temperature, relative humidity, and covariates and correcting for multiple testing. We also used independent component analysis (ICA) to examine the relative contribution of patterns of PM2.5 species. RESULTS Between 2000 and 2016, 456 men provided 648 blood samples, in which 1158 metabolites were quantified. We chose 305 metabolites for the short-term and 288 metabolites for the long-term exposure in this analysis that were significantly associated (p-value < 0.01) with PM2.5 to include in our PM2.5 species analysis. On average, men were 75.0 years old and their body mass index was 27.7 kg/m2. Only 3% were current smokers. In the adjusted models, ultrafine particles (UFPs) were the most significant species of short-term PM2.5 exposure followed by nickel, vanadium, potassium, silicon, and aluminum. Black carbon, vanadium, zinc, nickel, iron, copper, and selenium were the significant species of long-term PM2.5 exposure. We identified several metabolic pathways perturbed with PM2.5 species including glycerophospholipid, sphingolipid, and glutathione. These pathways are involved in inflammation, oxidative stress, immunity, and nucleic acid damage and repair. Results were overlapped with the ICA. CONCLUSIONS We identified several significant perturbed plasma metabolites and metabolic pathways associated with exposure to PM2.5 species. These species are associated with traffic, fuel oil, and wood smoke. This is the largest study to report a metabolomic signature of PM2.5 species' exposure and the first to use ICA.
Collapse
Affiliation(s)
- Feiby L Nassan
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA.
| | - Cuicui Wang
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Rachel S Kelly
- Channing Division of Network Medicine; Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine; Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Pantel S Vokonas
- VA Normative Aging Study, VA Boston Healthcare System, School of Medicine and School of Public Health, Boston University, Boston, MA 02215, USA
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Joel D Schwartz
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA; Channing Division of Network Medicine; Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02129, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
47
|
Mprah R, Adzika GK, Gyasi YI, Ndzie Noah ML, Adu-Amankwaah J, Adekunle AO, Duah M, Wowui PI, Weili Q. Glutaminolysis: A Driver of Vascular and Cardiac Remodeling in Pulmonary Arterial Hypertension. Front Cardiovasc Med 2021; 8:667446. [PMID: 33996951 PMCID: PMC8113389 DOI: 10.3389/fcvm.2021.667446] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a decimating ailment described by chronic precapillary pulmonary hypertension, an elevated mean pulmonary arterial pressure with a normal pulmonary capillary wedge pressure, and a raised pulmonary vascular resistance resulting in increased right ventricular afterload culminating in heart failure and death. Current PAH treatments regulate the vasodilatory/vasoconstrictory balance of pulmonary vessels. However, these treatment options are unable to stop the progression of, or reverse, an already established disease. Recent studies have advanced a metabolic dysregulation, featuring increased glutamine metabolism, as a mechanism driving PAH progression. Metabolic dysregulation in PAH leads to increased glutaminolysis to produce substrate to meet the high-energy requirement by hyperproliferative and apoptosis-resistant pulmonary vascular cells. This article explores the role of glutamate metabolism in PAH and how it could be targeted as an anti-remodeling therapeutic strategy.
Collapse
Affiliation(s)
- Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | - Yusif I. Gyasi
- Department of Chemistry & Biochemistry, Central Michigan University, Mount Pleasant, TX, United States
| | | | | | | | - Maxwell Duah
- Haematology Department, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | | | - Qiao Weili
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
48
|
Gallina AL, Rykaczewska U, Wirka RC, Caravaca AS, Shavva VS, Youness M, Karadimou G, Lengquist M, Razuvaev A, Paulsson-Berne G, Quertermous T, Gisterå A, Malin SG, Tarnawski L, Matic L, Olofsson PS. AMPA-Type Glutamate Receptors Associated With Vascular Smooth Muscle Cell Subpopulations in Atherosclerosis and Vascular Injury. Front Cardiovasc Med 2021; 8:655869. [PMID: 33959644 PMCID: PMC8093397 DOI: 10.3389/fcvm.2021.655869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/11/2021] [Indexed: 12/22/2022] Open
Abstract
Objectives and Aims: Vascular smooth muscle cells (VSMCs) are key constituents of both normal arteries and atherosclerotic plaques. They have an ability to adapt to changes in the local environment by undergoing phenotypic modulation. An improved understanding of the mechanisms that regulate VSMC phenotypic changes may provide insights that suggest new therapeutic targets in treatment of cardiovascular disease (CVD). The amino-acid glutamate has been associated with CVD risk and VSMCs metabolism in experimental models, and glutamate receptors regulate VSMC biology and promote pulmonary vascular remodeling. However, glutamate-signaling in human atherosclerosis has not been explored. Methods and Results: We identified glutamate receptors and glutamate metabolism-related enzymes in VSMCs from human atherosclerotic lesions, as determined by single cell RNA sequencing and microarray analysis. Expression of the receptor subunits glutamate receptor, ionotropic, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic (AMPA)-type subunit 1 (GRIA1) and 2 (GRIA2) was restricted to cells of mesenchymal origin, primarily VSMCs, as confirmed by immunostaining. In a rat model of arterial injury and repair, changes of GRIA1 and GRIA2 mRNA level were most pronounced at time points associated with VSMC proliferation, migration, and phenotypic modulation. In vitro, human carotid artery SMCs expressed GRIA1, and selective AMPA-type receptor blocking inhibited expression of typical contractile markers and promoted pathways associated with VSMC phenotypic modulation. In our biobank of human carotid endarterectomies, low expression of AMPA-type receptor subunits was associated with higher content of inflammatory cells and a higher frequency of adverse clinical events such as stroke. Conclusion: AMPA-type glutamate receptors are expressed in VSMCs and are associated with phenotypic modulation. Patients suffering from adverse clinical events showed significantly lower mRNA level of GRIA1 and GRIA2 in their atherosclerotic lesions compared to asymptomatic patients. These results warrant further mapping of neurotransmitter signaling in the pathogenesis of human atherosclerosis.
Collapse
Affiliation(s)
- Alessandro L Gallina
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Urszula Rykaczewska
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Robert C Wirka
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States
| | - April S Caravaca
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Vladimir S Shavva
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Mohamad Youness
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Glykeria Karadimou
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Mariette Lengquist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Anton Razuvaev
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Gabrielle Paulsson-Berne
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Thomas Quertermous
- Division of Cardiovascular Medicine and Cardiovascular Institute, School of Medicine, Stanford University, California, CA, United States
| | - Anton Gisterå
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Stephen G Malin
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Laura Tarnawski
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ljubica Matic
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Peder S Olofsson
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| |
Collapse
|
49
|
Deng P, Valentino T, Flythe MD, Moseley HNB, Leachman JR, Morris AJ, Hennig B. Untargeted Stable Isotope Probing of the Gut Microbiota Metabolome Using 13C-Labeled Dietary Fibers. J Proteome Res 2021; 20:2904-2913. [PMID: 33830777 DOI: 10.1021/acs.jproteome.1c00124] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The gut microbiome generates numerous metabolites that exert local effects and enter the circulation to affect the functions of many organs. Despite extensive sequencing-based characterization of the gut microbiome, there remains a lack of understanding of microbial metabolism. Here, we developed an untargeted stable isotope-resolved metabolomics (SIRM) approach for the holistic study of gut microbial metabolites. Viable microbial cells were extracted from fresh mice feces and incubated anaerobically with 13C-labeled dietary fibers including inulin or cellulose. High-resolution mass spectrometry was used to monitor 13C enrichment in metabolites associated with glycolysis, the Krebs cycle, the pentose phosphate pathway, nucleotide synthesis, and pyruvate catabolism in both microbial cells and the culture medium. We observed the differential use of inulin and cellulose as substrates for biosynthesis of essential and non-essential amino acids, neurotransmitters, vitamin B5, and other coenzymes. Specifically, the use of inulin for these biosynthetic pathways was markedly more efficient than the use of cellulose, reflecting distinct metabolic pathways of dietary fibers in the gut microbiome, which could be related with host effects. This technology facilitates deeper and holistic insights into the metabolic function of the gut microbiome (Metabolomic Workbench Study ID: ST001651).
Collapse
Affiliation(s)
- Pan Deng
- Superfund Research Center, University of Kentucky, Lexington 40536, Kentucky, United States.,Department of Pharmaceutical Sciences, University of Kentucky, Lexington 40536, Kentucky, United States
| | - Taylor Valentino
- Department of Physiology, University of Kentucky, Lexington 40536, Kentucky, United States
| | - Michael D Flythe
- Department of Animal and Food Sciences, University of Kentucky, Lexington 40536, Kentucky, United States.,United States Department of Agriculture, Agriculture Research Service, Forage Animal Production Research Unit, Lexington 40536, Kentucky, United States
| | - Hunter N B Moseley
- Superfund Research Center, University of Kentucky, Lexington 40536, Kentucky, United States.,Institute for Biomedical Informatics, University of Kentucky, Lexington 40536, Kentucky, United States.,Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington 40536, Kentucky, United States
| | - Jacqueline R Leachman
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington 40536, Kentucky, United States
| | - Andrew J Morris
- Superfund Research Center, University of Kentucky, Lexington 40536, Kentucky, United States.,Division of Cardiovascular Medicine, University of Kentucky, Lexington 40536, Kentucky, United States
| | - Bernhard Hennig
- Superfund Research Center, University of Kentucky, Lexington 40536, Kentucky, United States.,Department of Animal and Food Sciences, University of Kentucky, Lexington 40536, Kentucky, United States
| |
Collapse
|
50
|
Renick PJ, Mulgaonkar A, Co CM, Wu CY, Zhou N, Velazquez A, Pennington J, Sherwood A, Dong H, Castellino L, Öz OK, Tang L, Sun X. Imaging of Actively Proliferating Bacterial Infections by Targeting the Bacterial Metabolic Footprint with d-[5- 11C]-Glutamine. ACS Infect Dis 2021; 7:347-361. [PMID: 33476123 DOI: 10.1021/acsinfecdis.0c00617] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Since most d-amino acids (DAAs) are utilized by bacterial cells but not by mammalian eukaryotic hosts, recently DAA-based molecular imaging strategies have been extensively explored for noninvasively differentiating bacterial infections from the host's inflammatory responses. Given glutamine's pivotal role in bacterial survival, cell growth, biofilm formation, and even virulence, here we report a new positron emission tomography (PET) imaging approach using d-5-[11C]glutamine (d-[5-11C]-Gln) for potential clinical assessment of bacterial infection through a comparative study with its l-isomer counterpart, l-[5-11C]-Gln. In both control and infected mice, l-[5-11C]-Gln had substantially higher uptake levels than d-[5-11C]-Gln in most organs except the kidneys, showing the expected higher use of l-[5-11C]-Gln by mammalian tissues and more efficient renal excretion of d-[5-11C]-Gln. Importantly, our work demonstrates that PET imaging with d-[5-11C]-Gln is capable of detecting infections induced by both Escherichia coli (E. coli) and methicillin-resistant Staphylococcus aureus (MRSA) in a dual-infection murine myositis model with significantly higher infection-to-background contrast than with l-[5-11C]-Gln (in E. coli, 1.64; in MRSA, 2.62, p = 0.0004). This can be attributed to the fact that d-[5-11C]-Gln is utilized by bacteria while being more efficiently cleared from the host tissues. We confirmed the bacterial infection imaging specificity of d-[5-11C]-Gln by comparing its uptake in active bacterial infections versus sterile inflammation and with 2-deoxy-2-[18F]fluoroglucose ([18F]FDG). These results together demonstrate the translational potential of PET imaging with d-[5-11C]-Gln for the noninvasive detection of bacterial infectious diseases in humans.
Collapse
|