1
|
Chamika WAS, Ho TC, Park JS, Marasinghe CK, Je JY, Chun BS. Thermally optimized subcritical water hydrolysis for green extraction of bioactive compounds from sea cucumber Stichopus japonicus. Food Chem 2025; 484:144368. [PMID: 40262291 DOI: 10.1016/j.foodchem.2025.144368] [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: 11/21/2024] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
This study investigated the bioactive compounds in Stichopus japonicus extracts obtained using subcritical water hydrolysis (SWH) at nine temperatures (115 °C-235 °C at 15 °C intervals). Increasing temperature enhanced the extraction efficiency and decreased the average molecular weight, extracts had abundant amino acids and phenolic compounds. Maximum protein (832.66 ± 15.00 mg BSA/g) and polysachchrides (32.02 ± 0.88 mg glucose/g) contents were achieved at 175 °C, while the highest phenolic content (23.93 ± 0.16 mg GAE/g) and antioxidant activity were observed at 220 °C. The highest α-amylase (18.62 % ± 1.17 %) and α-glucosidase (24.31 % ± 1.43 %) activities were observed at 115 °C. However, there was no significant trend between temperature and anti-inflammatory or anticholesterol activities. GC-MS analysis confirmed the presence of bioactive compounds with antioxidant, anti-inflammatory, antidiabetic, and anticholesterol potential. In conclusion, our results indicate that SWH is a sustainable approach for extracting bioactive compounds from S. japonicus, with promising applications in functional foods and therapeutics.
Collapse
Affiliation(s)
- Weerathunga Arachchige Shiran Chamika
- Department of Food Science and Technology, Pukyong National University, 45 Yongso-ro, Nam-Gu, Busan 48513, Republic of Korea; Institute for Research & Development, 393/3, Lily Avenue, Robert Gunawardane Mawatha, Battaramulla, 10120, Sri Lanka
| | - Truc Cong Ho
- PL MICROMED Co., Ltd., 1F, 15-5, Yangju 3-gil, Yangsan-si, Gyeongsangnam-do 50620, Republic of Korea
| | - Jin-Seok Park
- Department of Food Science and Technology, Pukyong National University, 45 Yongso-ro, Nam-Gu, Busan 48513, Republic of Korea
| | | | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Byung-Soo Chun
- Department of Food Science and Technology, Pukyong National University, 45 Yongso-ro, Nam-Gu, Busan 48513, Republic of Korea.
| |
Collapse
|
2
|
Zeng X, Shi C, Han Y, Hu K, Li X, Wei C, Ding L, Cui J, Huang S, Xu Y, Zhang M, Shan W, Luo Q, Yu J, Zheng Z, Li X, Qian P, Huang H. A metabolic atlas of blood cells in young and aged mice identifies uridine as a metabolite to rejuvenate aged hematopoietic stem cells. NATURE AGING 2024; 4:1477-1492. [PMID: 39020094 DOI: 10.1038/s43587-024-00669-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/19/2024] [Indexed: 07/19/2024]
Abstract
Aging of hematopoietic stem cells (HSCs) is accompanied by impaired self-renewal ability, myeloid skewing, immunodeficiencies and increased susceptibility to malignancies. Although previous studies highlighted the pivotal roles of individual metabolites in hematopoiesis, comprehensive and high-resolution metabolomic profiles of different hematopoietic cells across ages are still lacking. In this study, we created a metabolome atlas of different blood cells across ages in mice. We reveal here that purine, pyrimidine and retinol metabolism are enriched in young hematopoietic stem and progenitor cells (HSPCs), whereas glutamate and sphingolipid metabolism are concentrated in aged HSPCs. Through metabolic screening, we identified uridine as a potential regulator to rejuvenate aged HSPCs. Mechanistically, uridine treatment upregulates the FoxO signaling pathway and enhances self-renewal while suppressing inflammation in aged HSCs. Finally, we constructed an open-source platform for public easy access and metabolomic analysis in blood cells. Collectively, we provide a resource for metabolic studies in hematopoiesis that can contribute to future anti-aging metabolite screening.
Collapse
Affiliation(s)
- Xiangjun Zeng
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Ce Shi
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Yingli Han
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Kejia Hu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Xiaoqing Li
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Cong Wei
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Lijuan Ding
- Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiazhen Cui
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Simao Huang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Yulin Xu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Meng Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Wei Shan
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Qian Luo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Jian Yu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | | | - Xia Li
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Hematology, Zhejiang University, Hangzhou, China.
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China.
| | - Pengxu Qian
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Hematology, Zhejiang University, Hangzhou, China.
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China.
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - He Huang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Hematology, Zhejiang University, Hangzhou, China.
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China.
| |
Collapse
|
3
|
Fang Y, Yin W, He C, Shen Q, Xu Y, Liu C, Zhou Y, Liu G, Zhao Y, Zhang H, Zhao K. Adverse impact of phthalate and polycyclic aromatic hydrocarbon mixtures on birth outcomes: A metabolome Exposome-Wide association study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 357:124460. [PMID: 38945193 DOI: 10.1016/j.envpol.2024.124460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
It has been well-investigating that individual phthalates (PAEs) or polycyclic aromatic hydrocarbons (PAHs) affect public health. However, there is still a gap that the mixture of PAEs and PAHs impacts birth outcomes. Through innovative methods for mixtures in epidemiology, we used a metabolome Exposome-Wide Association Study (mExWAS) to evaluate and explain the association between exposure to PAEs and PAHs mixtures and birth outcomes. Exposure to a higher level of PAEs and PAHs mixture was associated with lower birth weight (maximum cumulative effect: 143.5 g) rather than gestational age. Mono(2-ethlyhexyl) phthalate (MEHP) (posterior inclusion probability, PIP = 0.51), 9-hydroxyphenanthrene (9-OHPHE) (PIP = 0.53), and 1-hydroxypyrene (1-OHPYR) (PIP = 0.28) were identified as the most important compounds in the mixture. In mExWAS, we successfully annotated four overlapping metabolites associated with both MEHP/9-OHPHE/1-OHPYR and birth weight, including arginine, stearamide, Arg-Gln, and valine. Moreover, several lipid-related metabolism pathways, including fatty acid biosynthesis and degradation, alpha-linolenic acid, and linoleic acid metabolism, were disturbed. In summary, these findings may provide new insights into the underlying mechanisms by which PAE and PAHs affect fetal growth.
Collapse
Affiliation(s)
- Yiwei Fang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49, North Garden Road, Haidian district, Beijing, 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Wenjun Yin
- Wuhan Prevention and Treatment Center for Occupational Diseases, Wuhan, 430015, China
| | - Chao He
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiuzi Shen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying Xu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuanzhong Zhou
- School of Public Health, Zunyi Medical University, Zunyi, 563060, China
| | - Guotao Liu
- NHC Key Laboratory of Birth Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou, 450000, China
| | - Yun Zhao
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; NHC Key Laboratory of Birth Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou, 450000, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; NHC Key Laboratory of Birth Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou, 450000, China.
| |
Collapse
|
4
|
Gómez-Pascual A, Naccache T, Xu J, Hooshmand K, Wretlind A, Gabrielli M, Lombardo MT, Shi L, Buckley NJ, Tijms BM, Vos SJB, Ten Kate M, Engelborghs S, Sleegers K, Frisoni GB, Wallin A, Lleó A, Popp J, Martinez-Lage P, Streffer J, Barkhof F, Zetterberg H, Visser PJ, Lovestone S, Bertram L, Nevado-Holgado AJ, Gualerzi A, Picciolini S, Proitsi P, Verderio C, Botía JA, Legido-Quigley C. Paired plasma lipidomics and proteomics analysis in the conversion from mild cognitive impairment to Alzheimer's disease. Comput Biol Med 2024; 176:108588. [PMID: 38761503 DOI: 10.1016/j.compbiomed.2024.108588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative condition for which there is currently no available medication that can stop its progression. Previous studies suggest that mild cognitive impairment (MCI) is a phase that precedes the disease. Therefore, a better understanding of the molecular mechanisms behind MCI conversion to AD is needed. METHOD Here, we propose a machine learning-based approach to detect the key metabolites and proteins involved in MCI progression to AD using data from the European Medical Information Framework for Alzheimer's Disease Multimodal Biomarker Discovery Study. Proteins and metabolites were evaluated separately in multiclass models (controls, MCI and AD) and together in MCI conversion models (MCI stable vs converter). Only features selected as relevant by 3/4 algorithms proposed were kept for downstream analysis. RESULTS Multiclass models of metabolites highlighted nine features further validated in an independent cohort (0.726 mean balanced accuracy). Among these features, one metabolite, oleamide, was selected by all the algorithms. Further in-vitro experiments in rodents showed that disease-associated microglia excreted oleamide in vesicles. Multiclass models of proteins stood out with nine features, validated in an independent cohort (0.720 mean balanced accuracy). However, none of the proteins was selected by all the algorithms. Besides, to distinguish between MCI stable and converters, 14 key features were selected (0.872 AUC), including tTau, alpha-synuclein (SNCA), junctophilin-3 (JPH3), properdin (CFP) and peptidase inhibitor 15 (PI15) among others. CONCLUSIONS This omics integration approach highlighted a set of molecules associated with MCI conversion important in neuronal and glia inflammation pathways.
Collapse
Affiliation(s)
- Alicia Gómez-Pascual
- Department of Information and Communications Engineering Faculty of Informatics, University of Murcia, Murcia, Spain; Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Talel Naccache
- Department of Data Science, City University of London, United Kingdom
| | - Jin Xu
- Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | | | | | | | - Marta Tiffany Lombardo
- CNR Institute of Neuroscience, 20854, Vedano al Lambro, Italy; School of Medicine and Surgery, University of Milano-Bicocca, 20126, Italy
| | - Liu Shi
- Novo Nordisk Research Centre Oxford (NNRCO), Oxford, United Kingdom
| | - Noel J Buckley
- Department of Psychiatry, University of Oxford, United Kingdom; Kavli Institute for Nanoscience Discovery, Denmark
| | - Betty M Tijms
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Mara Ten Kate
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Department of Neurology and Bru-BRAIN, UZ Brussel and Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Kristel Sleegers
- Complex Genetics Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Giovanni B Frisoni
- University of Geneva, Geneva, Switzerland; IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Anders Wallin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Alberto Lleó
- Neurology Department, Hospital Sant Pau, Barcelona, Spain, Centro de Investigación en Red en enfermedades neurodegenerativas (CIBERNED)
| | - Julius Popp
- Old age psychiatry, University Hospital of Lausanne, University of Lausanne, Switzerland; Department of Geriatric Psychiatry, University Hospital of Psychiatry Zürich, University of Zürich, Switzerland
| | | | - Johannes Streffer
- AC Immune SA, Lausanne, Switzerland, formerly Janssen R&D, LLC. Beerse, Belgium at the time of study conduct
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, United Kingdom
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, United Kingdom; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Pieter Jelle Visser
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands; Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, United Kingdom; Janssen Medical (UK), High Wycombe, United Kingdom
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany; Department of Psychology, University of Oslo, Oslo, Norway
| | | | - Alice Gualerzi
- IRCCS Fondazione Don Carlo Gnocchi ONLUS in Milan, Italy
| | | | - Petroula Proitsi
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | | | - Juan A Botía
- Department of Information and Communications Engineering Faculty of Informatics, University of Murcia, Murcia, Spain
| | - Cristina Legido-Quigley
- Steno Diabetes Center Copenhagen, Herlev, Denmark; Institute of Pharmaceutical Science, King's College London, London, United Kingdom.
| |
Collapse
|
5
|
Yen NTH, Phat NK, Oh JH, Park SM, Moon KS, Thu VTA, Cho YS, Shin JG, Long NP, Kim DH. Pathway-level multi-omics analysis of the molecular mechanisms underlying the toxicity of long-term tacrolimus exposure. Toxicol Appl Pharmacol 2023; 473:116597. [PMID: 37321324 DOI: 10.1016/j.taap.2023.116597] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/17/2023]
Abstract
Tacrolimus (TAC)-based treatment is associated with nephrotoxicity and hepatotoxicity; however, the underlying molecular mechanisms responsible for this toxicity have not been fully explored. This study elucidated the molecular processes underlying the toxic effects of TAC using an integrative omics approach. Rats were sacrificed after 4 weeks of daily oral TAC administration at a dose of 5 mg/kg. The liver and kidney underwent genome-wide gene expression profiling and untargeted metabolomics assays. Molecular alterations were identified using individual data profiling modalities and further characterized by pathway-level transcriptomics-metabolomics integration analysis. Metabolic disturbances were mainly related to an imbalance in oxidant-antioxidant status, as well as in lipid and amino acid metabolism in the liver and kidney. Gene expression profiles also indicated profound molecular alterations, including in genes associated with a dysregulated immune response, proinflammatory signals, and programmed cell death in the liver and kidney. Joint-pathway analysis indicated that the toxicity of TAC was associated with DNA synthesis disruption, oxidative stress, and cell membrane permeabilization, as well as lipid and glucose metabolism. In conclusion, our pathway-level integration of transcriptome and metabolome and conventional analyses of individual omics profiles, provided a more comprehensive picture of the molecular changes resulting from TAC toxicity. This study also serves as a valuable resource for subsequent investigations aiming to understand the mechanism underlying the molecular toxicology of TAC.
Collapse
Affiliation(s)
- Nguyen Thi Hai Yen
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Ky Phat
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Jung-Hwa Oh
- Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Se-Myo Park
- Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Kyoung-Sik Moon
- Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Vo Thuy Anh Thu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Yong-Soon Cho
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Jae-Gook Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan 47392, Republic of Korea.
| | - Dong Hyun Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea.
| |
Collapse
|
6
|
Zhu Y, Xie Q, Ye J, Wang R, Yin X, Xie W, Li D. Metabolic Mechanism of Bacillus sp. LM24 under Abamectin Stress. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3068. [PMID: 36833759 PMCID: PMC9965259 DOI: 10.3390/ijerph20043068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
Abamectin (ABM) has been recently widely used in aquaculture. However, few studies have examined its metabolic mechanism and ecotoxicity in microorganisms. This study investigated the molecular metabolic mechanism and ecotoxicity of Bacillus sp. LM24 (B. sp LM24) under ABM stress using intracellular metabolomics. The differential metabolites most affected by the bacteria were lipids and lipid metabolites. The main significant metabolic pathways of B. sp LM24 in response to ABM stress were glycerolipid; glycine, serine, and threonine; and glycerophospholipid, and sphingolipid. The bacteria improved cell membrane fluidity and maintained cellular activity by enhancing the interconversion pathway of certain phospholipids and sn-3-phosphoglycerol. It obtained more extracellular oxygen and nutrients to adjust the lipid metabolism pathway, mitigate the impact of sugar metabolism, produce acetyl coenzyme A to enter the tricarboxylic acid (TCA) cycle, maintain sufficient anabolic energy, and use some amino acid precursors produced during the TCA cycle to express ABM efflux protein and degradative enzymes. It produced antioxidants, including hydroxyanigorufone, D-erythroascorbic acid 1'-a-D-xylopyranoside, and 3-methylcyclopentadecanone, to alleviate ABM-induced cellular and oxidative damage. However, prolonged stress can cause metabolic disturbances in the metabolic pathways of glycine, serine, threonine, and sphingolipid; reduce acetylcholine production; and increase quinolinic acid synthesis.
Collapse
Affiliation(s)
- Yueping Zhu
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Petrochemical Pollution Processes and Control, School of Environmental Science and Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China
| | - Qilai Xie
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agricultural and Pural Pullution Abatement and Environmental Safety, Guangzhou 510642, China
| | - Jinshao Ye
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Ruzhen Wang
- Guangdong Provincial Key Laboratory of Petrochemical Pollution Processes and Control, School of Environmental Science and Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China
| | - Xudong Yin
- Guangdong Provincial Key Laboratory of Petrochemical Pollution Processes and Control, School of Environmental Science and Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China
| | - Wenyu Xie
- Guangdong Provincial Key Laboratory of Petrochemical Pollution Processes and Control, School of Environmental Science and Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China
| | - Dehao Li
- Guangdong Provincial Key Laboratory of Petrochemical Pollution Processes and Control, School of Environmental Science and Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China
| |
Collapse
|
7
|
Yerlikaya S, Djamgoz MB. Oleamide, a Sleep-Inducing Compound: Effects on Ion Channels and Cancer. Bioelectricity 2022. [DOI: 10.1089/bioe.2022.0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Serife Yerlikaya
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Mustafa B.A. Djamgoz
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Biotechnology Research Center, Cyprus International University, Haspolat, Nicosia, TRNC, Mersin 10, Turkey
| |
Collapse
|
8
|
Yamamoto-Hijikata S, Suga K, Homareda H, Ushimaru M. Inhibition of the human secretory pathway Ca 2+, Mn 2+-ATPase1a by 1,3-thiazole derivatives. Biochem Biophys Res Commun 2022; 614:56-62. [PMID: 35567944 DOI: 10.1016/j.bbrc.2022.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/04/2022] [Indexed: 11/02/2022]
Abstract
The human Golgi/secretory pathway Ca2+,Mn2+-ATPase 1 (hSPCA1) transports Ca2+ and Mn2+ into the Golgi lumen. Studies of the biological functions of hSPCA1 are limited by a lack of selective pharmacological tools for SPCA1 inhibition. The aim of this study was therefore to identify compounds that specifically inhibit hSPCA1 activity. We found that five 1,3-thiazole derivatives exhibited inhibitory action towards the ATP hydrolysis activity of hSPCA1a in a concentration-dependent manner. Among the derivatives tested, compound 1 was the most potent, completely inhibiting hSPCA1a activity with a half-maximal inhibition (IC50) value of 0.8 μM. Compound 1 also partially inhibited the activity of another Ca2+,Mn2+-ATPase (hSPCA2) and Ca2+-ATPase (rSERCA1a), but had no effect on Na+,K+-ATPase or H+,K+-ATPase. Treatment of HeLa cells with compound 1 led to fragmentation of the Golgi ribbon into smaller stacks. In addition, compound 1 mobilized intracellular Ca2+ in HeLa cells that had been pre-treated with thapsigargin. Therefore, based on its selectivity and potency, compound 1 may be a valuable tool with which to further explore the role of SPCA1 in cellular processes.
Collapse
Affiliation(s)
| | - Kei Suga
- Department of Chemistry, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611, Japan
| | - Haruo Homareda
- Department of Chemistry, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611, Japan
| | - Makoto Ushimaru
- Department of Chemistry, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611, Japan
| |
Collapse
|
9
|
André R, Guedes R, López J, Serralheiro ML. Untargeted metabolomic study of HepG2 cells under the effect of Fucus vesiculosus aqueous extract. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e9197. [PMID: 34515383 DOI: 10.1002/rcm.9197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/28/2021] [Accepted: 09/09/2021] [Indexed: 06/13/2023]
Abstract
RATIONALE Fucus vesiculosus has been described with potential to develop functional foods containing bioactive compounds against various diseases. However, more studies are needed to better understand its functioning and its previously reported bioactivities, mainly at the molecular level. METHODS An untargeted metabolomic study was performed to analyse HepG2 cells exposed to F. vesiculosus aqueous extract, rich in phlorotannins and peptides, during 24 h. This study was carried out using liquid chromatography combined with high-resolution tandem mass spectrometry. RESULTS This metabolomic study showed significant changes in HepG2 metabolites in the presence of the extract, standing out being the increased intensity of various fatty acid amides (oleamide, (Z)-eicos-11-enamide, linoleamide, palmitamide, dodecanamide and stearamide). This group of metabolites is reported in the literature with anticancer and hypocholesterolemic activity, bioactivities also described for F. vesiculosus. The extract induced, likewise, the expression of glutathione indicating its antioxidant effect. CONCLUSIONS This study demonstrated the potential of the compounds present in the F. vesiculosus aqueous extract for the development of natural drugs, nutraceuticals or dietary supplements, justified at the molecular level by changes in cell metabolites related to anticancer and hypocholesterolemic activity. The results here described, using an untargeted metabolomic approach, may contribute to a better understanding of algal behaviour, when used as food, in health-promoting effects.
Collapse
Affiliation(s)
- Rebeca André
- Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, University of Lisbon, Campo Grande, Lisbon, Portugal
| | - Rita Guedes
- Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, University of Lisbon, Campo Grande, Lisbon, Portugal
| | - Javier López
- Parque Empresarial Rivas Futura, Bruker Española SA, Rivas Vaciamadrid, Madrid, Spain
| | - Maria Luísa Serralheiro
- Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, University of Lisbon, Campo Grande, Lisbon, Portugal
- Faculty of Sciences, Departamento de Química e Bioquímica, University of Lisbon, Campo Grande, Lisbon, Portugal
| |
Collapse
|
10
|
Oleamide Induces Cell Death in Glioblastoma RG2 Cells by a Cannabinoid Receptor-Independent Mechanism. Neurotox Res 2020; 38:941-956. [PMID: 32930995 DOI: 10.1007/s12640-020-00280-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/06/2020] [Accepted: 08/27/2020] [Indexed: 12/30/2022]
Abstract
The endocannabinoid system has been associated with antiproliferative effects in several types of tumors through cannabinoid receptor-mediated cell death mechanisms. Oleamide (ODA) is a CB1/CB2 agonist associated with cell growth and migration by adhesion and/or ionic signals associated with Gap junctions. Antiproliferative mechanisms related to ODA remain unknown. In this work, we evaluated the effects of ODA on cell viability and morphological changes in a rat RG2 glioblastoma cell line and compared these effects with primary astrocyte cultures from 8-day postnatal rats. RG2 and primary astrocyte cultures were treated with ODA at increasing concentrations (25, 50, 100, and 200 μM) for different periods of time (12, 24, and 48 h). Changes in RG2 cell viability and morphology induced by ODA were assessed by viability/mitochondrial activity test and phase contrast microscopy, respectively. The ratios of necrotic and apoptotic cell death, and cell cycle alterations, were evaluated by flow cytometry. The roles of CB1 and CB2 receptors on ODA-induced changes were explored with specific receptor antagonists. ODA (100 μM) induced somatic damage, detachment of somatic bodies, cytoplasmic polarization, and somatic shrinkage in RG2 cells at 24 and 48 h. In contrast, primary astrocytes treated at the same ODA concentrations exhibited cell aggregation but not cell damage. ODA (100 μM) increased apoptotic cell death and cell arrest in the G1 phase at 24 h in the RG2 line. The effects induced by ODA on cell viability of RG2 cells were independent of CB1 and CB2 receptors or changes in intracellular calcium transient. Results of this novel study suggest that ODA exerts specific antiproliferative effects on RG2 glioblastoma cells through unconventional apoptotic mechanisms not involving canonical signals.
Collapse
|
11
|
Adamska-Patruno E, Samczuk P, Ciborowski M, Godzien J, Pietrowska K, Bauer W, Gorska M, Barbas C, Kretowski A. Metabolomics Reveal Altered Postprandial Lipid Metabolism After a High-Carbohydrate Meal in Men at High Genetic Risk of Diabetes. J Nutr 2019; 149:915-922. [PMID: 31049566 DOI: 10.1093/jn/nxz024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/17/2018] [Accepted: 01/30/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The transcription factor 7-like 2 (TCF7L2) gene confers one of the strongest genetic predispositions to type 2 diabetes, but diabetes development can be modified by diet. OBJECTIVE The aim of our study was to evaluate postprandial metabolic alterations in healthy men with a high genetic risk of diabetes, after two meals with varying macronutrient content. METHODS The study was conducted in 21 homozygous nondiabetic men carrying the high-risk (HR, n = 8, age: 31.2 ± 6.3 y, body mass index (BMI, kg/m2) 28.5 ± 8.1) or low-risk (LR, n = 13, age: 35.2 ± 10.3 y, BMI: 28.1 ± 6.4) genotypes at the rs7901695 locus. During two meal challenge test visits subjects received standardized isocaloric (450 kcal) liquid meals: high-carbohydrate (HC, carbohydrates: 89% of energy) and normo-carbohydrate (NC, carbohydrates: 45% of energy). Fasting (0 min) and postprandial (30, 60, 120, 180 min) plasma samples were analyzed for metabolite profiles through untargeted metabolomics. Metabolic fingerprinting was performed on an ultra-high-performance liquid chromatography (UHPLC) system connected to an iFunnel quadrupole-time-of-flight (Q-TOF) mass spectrometer. RESULTS In HR-genotype men, after the intake of an HC-meal, we noted a significantly lower area under the curves (AUCs) of postprandial plasma concentrations of most of the phospholipids (-37% to -53%, variable importance in the projection (VIP) = 1.2-1.5), lysophospholipids (-29% to -86%, VIP = 1.1-2.6), sphingolipids (-32% to -47%, VIP = 1.1-1.3), as well as arachidonic (-36%, VIP = 1.4) and oleic (-63%, VIP = 1.3) acids, their metabolites: keto- and hydoxy-fatty acids (-38% to -78%, VIP = 1.3-2.5), leukotrienes (-65% to -83%, VIP = 1.4-2.2), uric acid (-59%, VIP = 1.5), and pyroglutamic acid (-65%, VIP = 1.8). The AUCs of postprandial sphingosine concentrations were higher (125-832%, VIP = 1.9-3.2) after the NC-meal, AUCs of acylcarnitines were lower (-21% to -61%, VIP = 1.1-2.4), and AUCs of fatty acid amides were higher (51-508%, VIP = 1.7-3.1) after the intake of both meals. CONCLUSIONS In nondiabetic men carrying the TCF7L2 HR genotype, subtle but detectable modifications in intermediate lipid metabolism are induced by an HC-meal. This trial was registered at www.clinicaltrials.gov as NCT03792685.
Collapse
Affiliation(s)
| | - Paulina Samczuk
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Joanna Godzien
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland.,Center for Metabolomics and Bioanalysis (CEMBIO), Universidad CEU San Pablo, Madrid, Spain
| | - Karolina Pietrowska
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Witold Bauer
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Maria Gorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Coral Barbas
- Center for Metabolomics and Bioanalysis (CEMBIO), Universidad CEU San Pablo, Madrid, Spain
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland.,Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
12
|
Adamska-Patruno E, Godzien J, Ciborowski M, Samczuk P, Bauer W, Siewko K, Gorska M, Barbas C, Kretowski A. The Type 2 Diabetes Susceptibility PROX1 Gene Variants Are Associated with Postprandial Plasma Metabolites Profile in Non-Diabetic Men. Nutrients 2019; 11:nu11040882. [PMID: 31010169 PMCID: PMC6520869 DOI: 10.3390/nu11040882] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/04/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
The prospero homeobox 1 (PROX1) gene may show pleiotropic effects on metabolism. We evaluated postprandial metabolic alterations dependently on the rs340874 genotypes, and 28 non-diabetic men were divided into two groups: high-risk (HR)-genotype (CC-genotype carriers, n = 12, 35.3 ± 9.5 years old) and low-risk (LR)-genotype (allele T carriers, n = 16, 36.3 ± 7.0 years old). Subjects participated in two meal-challenge-tests with high-carbohydrate (HC, carbohydrates 89%) and normo-carbohydrate (NC, carbohydrates 45%) meal intake. Fasting and 30, 60, 120, and 180 min after meal intake plasma samples were fingerprinted by liquid chromatography quadrupole time-of-flight mass spectrometry (LC-QTOF-MS). In HR-genotype men, the area under the curve (AUC) of acetylcarnitine levels was higher after the HC-meal [+92%, variable importance in the projection (VIP) = 2.88] and the NC-meal (+55%, VIP = 2.00) intake. After the NC-meal, the HR-risk genotype carriers presented lower AUCs of oxidized fatty acids (−81–66%, VIP = 1.43–3.16) and higher linoleic acid (+80%, VIP = 2.29), while after the HC-meal, they presented lower AUCs of ornithine (−45%, VIP = 1.83), sphingosine (−48%, VIP = 2.78), linoleamide (−45%, VIP = 1.51), and several lysophospholipids (−40–56%, VIP = 1.72–2.16). Moreover, lower AUC (−59%, VIP = 2.43) of taurocholate after the HC-meal and higher (+70%, VIP = 1.42) glycodeoxycholate levels after the NC-meal were observed. Our results revealed differences in postprandial metabolites from inflammatory and oxidative stress pathways, bile acids signaling, and lipid metabolism in PROX1 HR-genotype men. Further investigations of diet–genes interactions by which PROX1 may promote T2DM development are needed.
Collapse
Affiliation(s)
- Edyta Adamska-Patruno
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland.
| | - Joanna Godzien
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland.
| | - Michal Ciborowski
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland.
| | - Paulina Samczuk
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland.
| | - Witold Bauer
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland.
| | - Katarzyna Siewko
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland.
| | - Maria Gorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland.
| | - Coral Barbas
- Center for Metabolomics and Bioanalysis (CEMBIO), Universidad CEU San Pablo, 28003 Madrid, Spain.
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland.
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland.
| |
Collapse
|
13
|
蒋 国, 刘 亚, 赵 婉, 王 道, 董 淑, 童 旭. [Effect of gap junction modulation on antitumor effects of adriamycin in estrogen receptor-positive breast cancer cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:780-786. [PMID: 33168517 PMCID: PMC6765543 DOI: 10.3969/j.issn.1673-4254.2018.07.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To observe the effect of functional modulation of gap junctions (GJ) on the antitumor effect of adriamycin in breast cancer cells positive for estrogen receptor (ER). METHODS The inhibitory effect of 0 to 24.0 μmol/L adriamycin on the surviving fraction of ER-positive human breast cancer MCF-7 cells and ER-negative MDA-MB-231 cells was assessed with MTT assay; Western blotting and immunofluorescence assay were used to detect the expressions of Cx43 total protein and membrane protein in the cells. A parachute assay was used to evaluate the function of the GJ in MCF-7 cells. The cytotoxic effect of adriamycin was observed in the cells treated with retinoic acid (RA) for enhancing GJ function, in cells treated with oleamide and 18-α- glycyrrhizic acid (18-α-ga) for inhibiting GJ function, and also in cells transfected with Cx43siRNA for Cx43 knockdown. RESULTS ER-positive MCF-7 cells expressed a significantly higher level of Cx43 with stronger GJ function than ER-negative MDA- MB-231 cells. Adriamycin significantly inhibited the proliferation of MCF-7 cells (P < 0.01), and RA treatment further increased the cytotoxicity of adriamycin (P < 0.01) while oleamide and 18-α-GA obviously attenuated the cytotoxicity of adriamycin (P < 0.01). In the cells with Cx43 knockdown, the expressions of total Cx43 protein and Cx43 on the membrane were significantly reduced, the function of GJ was attenuated, and the cytotoxicity of adriamycin was significantly decreased (P < 0.01). CONCLUSIONS ER-positive breast cancer cells have stronger Cx43 expressions and GJ function than the ERnegative cells. The cytotoxicity of adriamycin against the breast cancer cells can be strengthened by enhancing GJ function and attenuated by inhibiting GJ function. Cx43 silencing inhibits the function of GJ to lower the cytotoxicity of adriamycin in human breast cancer MCF-7 cells.
Collapse
Affiliation(s)
- 国君 蒋
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| | - 亚明 刘
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| | - 婉晨 赵
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| | - 道鑫 王
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| | - 淑英 董
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| | - 旭辉 童
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| |
Collapse
|
14
|
Abdullah M, Kornegay JN, Honcoop A, Parry TL, Balog-Alvarez CJ, O'Neal SK, Bain JR, Muehlbauer MJ, Newgard CB, Patterson C, Willis MS. Non-Targeted Metabolomics Analysis of Golden Retriever Muscular Dystrophy-Affected Muscles Reveals Alterations in Arginine and Proline Metabolism, and Elevations in Glutamic and Oleic Acid In Vivo. Metabolites 2017; 7:E38. [PMID: 28758940 PMCID: PMC5618323 DOI: 10.3390/metabo7030038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/21/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Like Duchenne muscular dystrophy (DMD), the Golden Retriever Muscular Dystrophy (GRMD) dog model of DMD is characterized by muscle necrosis, progressive paralysis, and pseudohypertrophy in specific skeletal muscles. This severe GRMD phenotype includes moderate atrophy of the biceps femoris (BF) as compared to unaffected normal dogs, while the long digital extensor (LDE), which functions to flex the tibiotarsal joint and serves as a digital extensor, undergoes the most pronounced atrophy. A recent microarray analysis of GRMD identified alterations in genes associated with lipid metabolism and energy production. METHODS We, therefore, undertook a non-targeted metabolomics analysis of the milder/earlier stage disease GRMD BF muscle versus the more severe/chronic LDE using GC-MS to identify underlying metabolic defects specific for affected GRMD skeletal muscle. RESULTS Untargeted metabolomics analysis of moderately-affected GRMD muscle (BF) identified eight significantly altered metabolites, including significantly decreased stearamide (0.23-fold of controls, p = 2.89 × 10-3), carnosine (0.40-fold of controls, p = 1.88 × 10-2), fumaric acid (0.40-fold of controls, p = 7.40 × 10-4), lactamide (0.33-fold of controls, p = 4.84 × 10-2), myoinositol-2-phosphate (0.45-fold of controls, p = 3.66 × 10-2), and significantly increased oleic acid (1.77-fold of controls, p = 9.27 × 10-2), glutamic acid (2.48-fold of controls, p = 2.63 × 10-2), and proline (1.73-fold of controls, p = 3.01 × 10-2). Pathway enrichment analysis identified significant enrichment for arginine/proline metabolism (p = 5.88 × 10-4, FDR 4.7 × 10-2), where alterations in L-glutamic acid, proline, and carnosine were found. Additionally, multiple Krebs cycle intermediates were significantly decreased (e.g., malic acid, fumaric acid, citric/isocitric acid, and succinic acid), suggesting that altered energy metabolism may be underlying the observed GRMD BF muscle dysfunction. In contrast, two pathways, inosine-5'-monophosphate (VIP Score 3.91) and 3-phosphoglyceric acid (VIP Score 3.08) mainly contributed to the LDE signature, with two metabolites (phosphoglyceric acid and inosine-5'-monophosphate) being significantly decreased. When the BF and LDE were compared, the most significant metabolite was phosphoric acid, which was significantly less in the GRMD BF compared to control and GRMD LDE groups. CONCLUSIONS The identification of elevated BF oleic acid (a long-chain fatty acid) is consistent with recent microarray studies identifying altered lipid metabolism genes, while alterations in arginine and proline metabolism are consistent with recent studies identifying elevated L-arginine in DMD patient sera as a biomarker of disease. Together, these studies demonstrate muscle-specific alterations in GRMD-affected muscle, which illustrate previously unidentified metabolic changes.
Collapse
Affiliation(s)
- Muhammad Abdullah
- Department of Biochemistry, QuaidiAzam University, 45320 Islamabad, Pakistan.
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525, USA.
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599-7126, USA.
| | - Joe N Kornegay
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA.
| | - Aubree Honcoop
- Toxicology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Traci L Parry
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525, USA.
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599-7126, USA.
| | - Cynthia J Balog-Alvarez
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA.
| | - Sara K O'Neal
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27708, USA.
| | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27708, USA.
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC 27703, USA.
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27708, USA.
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27708, USA.
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC 27703, USA.
| | - Cam Patterson
- Presbyterian Hospital/Weill-Cornell Medical Center, New York, NY 10065, USA.
| | - Monte S Willis
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525, USA.
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599-7126, USA.
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|