1
|
Hanaoka C, Pichika R, Dayanidhi S, Jayabalan P. Serum metabolomics after exercise in ambulatory individuals with cerebral palsy. Dev Med Child Neurol 2025; 67:639-647. [PMID: 39431769 PMCID: PMC11965978 DOI: 10.1111/dmcn.16105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 07/31/2024] [Accepted: 08/20/2024] [Indexed: 10/22/2024]
Abstract
AIM To evaluate whether serum metabolomics differ between ambulatory individuals with cerebral palsy (CP) compared with individuals with typical development and whether functional capacity is associated with metabolite abundance. METHOD Thirty-eight adolescents and young adults were enrolled (CP: n = 19; typical development: n = 19). After functional capacity testing (10-meter walk, sit-to-stand, and peak knee flexion/extension torques), blood was drawn. Targeted serum metabolomics on hydrophilic metabolites were performed by high-performance liquid chromatography coupled with high-resolution and tandem mass spectrometry. Metabolite dimensionality reduction, pathway analysis, fold change, and t-tests evaluated changes in metabolite abundance. Associations were tested between functional measures and metabolite abundance. RESULTS Individuals with CP had a significant increase in the abundance of essential amino acids, catabolic products of protein metabolism, and tricarboxylic acid cycle substrates, such as valine, tryptophan, kynurenic acid, and pyruvate (p < 0.05). Importantly, the abundance of numerous metabolites was only highly associated with functional capacity in individuals with CP such that greater abundance was associated with greater capacity, but not in those with typical development. INTERPRETATION Our findings show clear increases in serum metabolites in individuals with CP, which are associated with functional capacity for movement. The altered metabolite profile measured after exercise might reflect increased energy production needed for movement. Appropriate nutritional intake during exercise might be needed given increased energy requirements.
Collapse
Affiliation(s)
- Chad Hanaoka
- Northwestern University Feinberg School of MedicineChicagoILUSA
- Shirley Ryan AbilityLabChicagoILUSA
| | - Rajeswari Pichika
- Northwestern University Feinberg School of MedicineChicagoILUSA
- Shirley Ryan AbilityLabChicagoILUSA
- Edward Hines VA Medical CenterHinesILUSA
| | - Sudarshan Dayanidhi
- Northwestern University Feinberg School of MedicineChicagoILUSA
- Shirley Ryan AbilityLabChicagoILUSA
| | - Prakash Jayabalan
- Northwestern University Feinberg School of MedicineChicagoILUSA
- Shirley Ryan AbilityLabChicagoILUSA
| |
Collapse
|
2
|
Ren M, Wang Y, Yuan Y, Du H, Liang Q, Qin F, Xiong Z. Integration of UHPLC-MS and mass spectrometry imaging techniques revealed the protective mechanism of Gushudan in postmenopausal osteoporosis rats via branched-chain amino acid metabolism based on the 'kidney-bone' axis. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1256:124540. [PMID: 40023006 DOI: 10.1016/j.jchromb.2025.124540] [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: 01/19/2025] [Revised: 02/09/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
According to the theory of traditional Chinese medicine, the kidney is regarded as governing the bones and dominating the storage of essence. Gushudan (GSD) is a traditional Chinese medicine prescription that has the effects of strengthening bone and nourishing the kidney. However, the mechanism of action of GSD in preventing postmenopausal osteoporosis (PMOP) rats based on the 'kidney-bone' axis remains to be further systematically investigated. In this study, an integrated kidney metabolomics method based on three MS detection modes of UHPLC-HRMS, UHPLC-MS/MS and AFADESI-MSI was developed to reveal the protective mechanism of GSD in PMOP rats. Firstly, the non-targeted metabolomics was investigated to comprehensively explore the metabolic changes in the kidneys of PMOP rats based on the UHPLC-Q-Orbitrap HRMS. Subsequently, UHPLC-MS/MS targeted metabolomics and Mass Spectrometry Imaging (MSI) techniques were combined to elucidate the preventive mechanism of GSD on PMOP through branched-chain amino acid (BCAA) metabolism. The results of the non-targeted metabolomics demonstrated that GSD significantly modulated the levels of 67 potential biomarkers, including leucine and valine, which are primarily involved in amino acid metabolism. Specifically, BCAA metabolism is notably enriched in amino acid metabolism. Compared to the control group, it was found that the levels of BCAAs were decreased and α-branched-chain keto acids (BCKAs) were increased in the model groups in the targeted metabolomics study. Moreover, MSI results showed that the changes in BCAAs content were mainly concentrated in the renal cortex. This finding confirmed the metabolic disorders of BCAA in the renal cortex of PMOP rats, and that GSD had a significant regulatory effect on this disorder. In conclusion, this study integrated three mass spectrometry techniques that validate and complement each other to revealed the anti-osteoporostic mechanism of GSD in PMOP rats and to elucidate the modern scientific connotation of the 'kidney-bone' axis based on the BCAA metabolism.
Collapse
Affiliation(s)
- Mengxin Ren
- School of Pharmacy, Shenyang Pharmaceutical University, No.26 Huatuo Rd, High & New Tech Development Zone, 117004 Benxi, Liaoning Province, PR China
| | - Yajing Wang
- School of Pharmacy, Shenyang Pharmaceutical University, No.26 Huatuo Rd, High & New Tech Development Zone, 117004 Benxi, Liaoning Province, PR China
| | - Yue Yuan
- School of Pharmacy, Shenyang Pharmaceutical University, No.26 Huatuo Rd, High & New Tech Development Zone, 117004 Benxi, Liaoning Province, PR China
| | - Hailing Du
- School of Pharmacy, Shenyang Pharmaceutical University, No.26 Huatuo Rd, High & New Tech Development Zone, 117004 Benxi, Liaoning Province, PR China
| | - Qinghua Liang
- School of Pharmacy, Shenyang Pharmaceutical University, No.26 Huatuo Rd, High & New Tech Development Zone, 117004 Benxi, Liaoning Province, PR China
| | - Feng Qin
- School of Pharmacy, Shenyang Pharmaceutical University, No.26 Huatuo Rd, High & New Tech Development Zone, 117004 Benxi, Liaoning Province, PR China
| | - Zhili Xiong
- School of Pharmacy, Shenyang Pharmaceutical University, No.26 Huatuo Rd, High & New Tech Development Zone, 117004 Benxi, Liaoning Province, PR China.
| |
Collapse
|
3
|
Yang SJ, Yu XK, Zuo Q. Branched- Chain Fatty Acids and Obesity: A Narrative Review. Nutr Rev 2025:nuaf022. [PMID: 40207993 DOI: 10.1093/nutrit/nuaf022] [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] [Indexed: 04/11/2025] Open
Abstract
Branched- chain fatty acids (BCFAs) are a category of saturated fatty acids that are commonly present in various organisms and play a crucial role in a variety of metabolic reactions, including anticancer, lipid-lowering, anti-inflammatory, and neuroprotective actions. Currently, there is growing interest in the relationship between BCFAs and obesity. Branched- chain fatty acids regulate the gene expression of related enzymes by activating PPARα and sterol regulatory element-binding protein-1c, thereby reducing triglyceride synthesis in the body. Additionally, BCFAs reduce inflammation by decreasing the expression of pro-inflammatory factors in obesity such as cyclooxygenase-2, interleukin-6, and lipoxygenase-15 genes. Branched- chain fatty acids can also expedite the conversion of branched chain amino acids to BCFAs to regulate obesity-induced insulin resistance. In this article we provide a comprehensive review of research progress on how BCFAs affect obesity from the perspectives of lipid metabolism, inflammation, and insulin resistance.
Collapse
Affiliation(s)
- Shi-Jiao Yang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xin-Kai Yu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Qun Zuo
- School of Sports Performance, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
4
|
Zhan C, Quan Z, Huang X, Bu J, Li S. Causal relationships of circulating amino acids with sarcopenia-related traits: A bidirectional Mendelian randomization study. Clin Nutr 2025; 47:258-264. [PMID: 40073510 DOI: 10.1016/j.clnu.2025.02.020] [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: 08/11/2024] [Revised: 01/26/2025] [Accepted: 02/16/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND AND AIM Recent studies have indicated a correlation between certain Amino acids (AAs) and sarcopenia. However, the exact causal relationship among these associations is still unclear. This study aims to elucidate the causal relationships between 20 types of AAs and the phenotypic characteristics associated with sarcopenia through Mendelian randomization (MR) analysis. METHODS AND RESULTS This MR study employed single nucleotide polymorphisms (SNPs) that were significantly associated with both AAs and the traits of sarcopenia as instrumental variables (IVs). The main method for estimating causal effects was the inverse-variance weighted (IVW) approach. To ensure the robustness of the findings, additional methods such as weighted median, weighted mode, and MR Egger regression were used. Sensitivity analyses included heterogeneity and pleiotropy tests. In this research, we discovered potential causal relationships between AAs and traits associated with sarcopenia. We not only found that AAs previously studied, such as Glutamine, Tyrosine, Glycine, and branched-chain amino acids, play positive roles in muscle metabolism. Additionally, our study identified the role of AAs previously neglected or not considered in earlier research, such as Alanine, Lysine, Cysteine, and Methionine, which exert potential effects on muscle metabolism and offer considerable research potential and value. CONCLUSIONS This MR study clarified the reciprocal effects between circulating levels of AAs and sarcopenia-related traits. These results indicate that AAs may be used as biomarkers for diagnosing sarcopenia or as intervention targets for its treatment in clinical practice.
Collapse
Affiliation(s)
- Chenyang Zhan
- Department of General Surgery, Chengdu Second People's Hospital, Chengdu 610041, China; School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
| | - Zongjie Quan
- Department of General Surgery, Chengdu Second People's Hospital, Chengdu 610041, China; School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Xiujin Huang
- Department of General Surgery, Chengdu Second People's Hospital, Chengdu 610041, China; School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Jun Bu
- Department of General Surgery, Chengdu Second People's Hospital, Chengdu 610041, China.
| | - Sheng Li
- Department of General Surgery, Chengdu Second People's Hospital, Chengdu 610041, China; School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
| |
Collapse
|
5
|
He X, Tinghäll Nilsson U, Mishchuk DO, Hernell O, Lönnerdal B, Hartvigsen ML, Jacobsen LN, Kvistgaard AS, Slupsky CM, Karlsland Åkeson P. Impact of formula protein quantity and source on infant metabolism: serum, urine, and fecal metabolomes of a randomized controlled study. Am J Clin Nutr 2025; 121:853-864. [PMID: 39921093 DOI: 10.1016/j.ajcnut.2025.02.002] [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: 09/17/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Human milk offers significant health benefits for infants; however, when not feasible, infant formula serves as an alternative. The higher protein content in infant formula is thought to contribute to the distinct metabolic profiles observed in formula-fed infants compared with those fed human milk. OBJECTIVES This study investigates the impact of formula protein quantity and whey protein types on the serum, urine, and fecal metabolomes of infants. METHODS A secondary analysis was performed on a random subset of 200 well-characterized per-protocol infants who completed a prospective, randomized, double-blind controlled trial. Infants were randomly assigned to 1 of 3 groups: standard formula, protein-reduced formula with α-lactalbumin-enriched whey, or protein-reduced formula with casein glycomacropeptide-reduced whey, along with an observational reference group of exclusively breastfed infants. Serum, urine, and fecal metabolites were quantified using 1H nuclear magnetic resonance spectroscopy at baseline (1-2 mo), 4, and 6 mo of age. Dietary intake was assessed monthly ≤6 mo of age. RESULTS Formula protein content and type of whey protein used significantly influenced the amino acid profile and associated catabolic markers in serum and urine but had minimal impact on the fecal metabolome. Reduced protein formulas yielded metabolome profiles closer to those of breastfed infants compared with standard formula. Despite these improvements, infants fed human milk still demonstrated enhanced branched-chain amino acid (BCAA) oxidation and a greater capacity to eliminate catabolic waste products from BCAA metabolism over infants consuming protein-reduced formulas. CONCLUSIONS Comprehensive metabolomics profiling of serum, urine, and feces captures molecular-level changes and informs potential strategies for formula optimization. Both the quantity and source of protein significantly influenced the metabolic profiles of formula-fed infants. However, modifications in protein alone cannot fully resolve the metabolic differences between formula-fed and breastfed infants, highlighting the complexity of mimicking the human milk feeding-associated metabolic profile. This study was registered at clinicaltrials.gov as NCT02410057.
Collapse
Affiliation(s)
- Xuan He
- Department of Nutrition, University of California-Davis, Davis, CA, United States; Department of Food Science and Technology, University of California-Davis, Davis, CA, United States
| | | | - Darya O Mishchuk
- Department of Food Science and Technology, University of California-Davis, Davis, CA, United States
| | - Olle Hernell
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Bo Lönnerdal
- Department of Nutrition, University of California-Davis, Davis, CA, United States
| | | | | | | | - Carolyn M Slupsky
- Department of Nutrition, University of California-Davis, Davis, CA, United States; Department of Food Science and Technology, University of California-Davis, Davis, CA, United States.
| | - Pia Karlsland Åkeson
- Department of Clinical Sciences Malmö, Pediatrics, Lund University, Malmö, Sweden
| |
Collapse
|
6
|
Fedorov VB, Garreau A, Tøien Ø, Barnes BM, Goropashnaya AV. Transcriptome remodeling and adaptive preservation of muscle protein content in hibernating black bears. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641932. [PMID: 40166218 PMCID: PMC11956910 DOI: 10.1101/2025.03.06.641932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Hibernation is an energy-saving adaptation associated with physical inactivity. In contrast to most mammals, hibernating bears demonstrate limited loss of muscle mass and protein content over the prolonged periods of immobility and fasting during winter. This suggests that bears have natural adaptive mechanisms preserving muscle mass and functionality. To identify transcriptional changes that underlie molecular mechanisms attenuating muscle loss, we conducted a large-scale gene expression profiling (14,199 genes) by transcriptome sequencing in quadriceps of adult black bears, comparing hibernating animals (n=5) and summer active animals (n=5). Gene set enrichment analysis showed significant positive correlation between hibernating phenotype and expression changes of genes involved in translation, ribosome and the mTORC1 mediated signaling. In contrast, coordinated transcriptional reduction was detected for genes involved in catabolism of branched chain amino acid (BCAA) suggesting preservation of BCAA. These findings imply maintenance of protein biosynthesis through the mTORC1 signaling positively activated by availability of BCAA in muscle during hibernation. Support for this conclusion comes from overexpression of RRAGD and RRAGB , crucial regulator of the mTORC1 response to leucine availability, and up regulation of EIF4B , downstream target of the mTORC1 signaling. Consistent with the mTORC1 suppression of autophagy-dependent protein degradation, MAP1LC3A and ULK1 were down regulated in hibernating muscle. The maintenance of protein biosynthesis and decrease in protein catabolism through the mTORC1 signaling as response to BCAA availability likely contribute to the preservation of muscle protein through prolonged periods of immobility and fasting during hibernation.
Collapse
|
7
|
Yeo M, Rehsi P, Yeo JM, Dixon M, Chakrapani A. Single centre retrospective review of plasma branched-chain amino acid levels in children with urea cycle disorders: Impact of treatment modalities and disease severity. Mol Genet Metab Rep 2025; 42:101190. [PMID: 39897473 PMCID: PMC11786853 DOI: 10.1016/j.ymgmr.2025.101190] [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: 09/24/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Branched-chain amino acids (BCAAs) are important for normal growth, development, and function. In urea cycle disorders (UCDs), plasma BCAA levels can be relatively low; this has been attributed variously to low protein intake, hyperammonaemia, and nitrogen scavenger treatment. We undertook a retrospective review of plasma BCAA levels in individuals with UCDs comprising ornithine carbamoyltransferase deficiency (OTCD n = 22), arginosuccinate lyase deficiency (ASLD n = 12), and argininosuccinate synthase deficiency (ASSD n = 6). Scavenger treatment groups comprised sodium benzoate (NaBz, n = 20), sodium phenylbutyrate (NaPBA, n = 5), NaBz+NaPBA (n = 14), and a control group receiving neither NaBz nor NaPBA (n = 14). In these treatment groups, respectively, median (IQR) plasma levels of leucine were 54 (32), 55 (25), 58 (19), and 91 (70) μmol/L (leucine was lower in the NaBz group than the control, p = 0.0282) and numbers of individuals (%) with leucine below normal were 10/20 (50 %), 1/4 (25 %), 10/14 (71 %), and 2/9 (20 %). The pattern was similar for isoleucine and valine. In the NaBz group, plasma BCAA levels were inversely correlated with protein intake (p ≤ 0.01 to p ≤ 0.001), plasma ammonia level (p ≤ 0.01 to p ≤ 0.0001), and scavenger dose (p ≤ 0.0001). We speculate that individuals with greater disease severity may be prone to BCAA deficiency, caused by BCAA consumption when alternative urea disposal pathways are used. Practical reflections on our audit were that to increase the proportion of plasma BCAA levels in the normal range, we needed to alter the biological value of protein intake, prescribe higher doses of scavenger to facilitate safe levels of protein intake, and give EAA supplements if indicated.
Collapse
Affiliation(s)
- Mildrid Yeo
- Department of Paediatric Inherited Metabolic Disease, Great Ormond Street Hospital for Children, NHS Foundation Trust and Institute for Child Health, London, UK
| | - Preeya Rehsi
- Department of Paediatric Inherited Metabolic Disease, Great Ormond Street Hospital for Children, NHS Foundation Trust and Institute for Child Health, London, UK
| | - Jie Ming Yeo
- Department of Paediatric Inherited Metabolic Disease, Great Ormond Street Hospital for Children, NHS Foundation Trust and Institute for Child Health, London, UK
| | - Marjorie Dixon
- Dietetics, Great Ormond Street Hospital for Children, NHS Foundation Trust and Institute for Child Health, London, UK
| | - Anupam Chakrapani
- Department of Paediatric Inherited Metabolic Disease, Great Ormond Street Hospital for Children, NHS Foundation Trust and Institute for Child Health, London, UK
| |
Collapse
|
8
|
Demianchuk O, Bayliak M, Vatashchuk M, Gospodaryov D, Hurza V, Derkachov V, Berezovskyi V, Lushchak VI. Alpha-ketoglutarate promotes anxiety, activates autophagy, and suppresses antioxidant enzymes in the cerebral cortex of female mice on cafeteria diet. Brain Res Bull 2025; 222:111255. [PMID: 39952443 DOI: 10.1016/j.brainresbull.2025.111255] [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: 10/02/2024] [Revised: 02/03/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025]
Abstract
Alpha-ketoglutarate (AKG), an intermediate of the tricarboxylic acid cycle, has been found to mitigate oxidative stress and inflammation. In turn, a cafeteria diet (CD), an obesogenic diet, is often associated with oxidative stress and inflammation. This study aimed to determine whether AKG can level the effects of CD on animal behavior, oxidative stress markers, glycolytic flow, and autophagy in the mouse cerebral cortex. Female C57BL/6 J mice were divided into two groups and fed either a standard diet or a CD for eight weeks. For the next four weeks, each group continued to be fed the previous diet; however, half of the individuals within each group received drinking water with 1 % AKG. Using an open field test, we found that the combination of CD and AKG promoted the development of anxiety signs. Both CD and AKG decreased the exploratory behavior of mice, with a significant additive effect in the combined diet. On diets supplemented with AKG, animals produced fewer fecal boli, a measure of emotionality. On all experimental diets, mice had lower activities of antioxidant and related enzymes, with no significant differences in the activities of glycolytic enzymes. The AKG-supplemented diet induced the transcription of autophagy-related genes and targets of the forkhead box O factor, involved in the regulation of carbohydrate metabolism. Transcriptional changes induced by AKG were partly abrogated by the CD. These findings suggest that AKG, particularly when combined with CD, may modulate behavioral responses and oxidative stress intensity in the brain by altering key metabolic and autophagic pathways.
Collapse
Affiliation(s)
- Oleh Demianchuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Maria Bayliak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine.
| | - Myroslava Vatashchuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Dmytro Gospodaryov
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine.
| | - Viktoriia Hurza
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Vitalii Derkachov
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Vladyslav Berezovskyi
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Volodymyr I Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine; Research and Development University, 13a Shota Rustaveli Str., Ivano-Frankivsk 76018, Ukraine.
| |
Collapse
|
9
|
Sadeesh EM, Lahamge MS. Unveiling the tissue-specific landscape of nuclear-encoded mitochondrial genes involved in amino acid metabolism in buffalo. Amino Acids 2025; 57:17. [PMID: 40019559 PMCID: PMC11870903 DOI: 10.1007/s00726-025-03447-4] [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: 10/16/2024] [Accepted: 02/16/2025] [Indexed: 03/01/2025]
Abstract
Mitochondria play a pivotal role in energy production, metabolism, and cellular signaling, serving as key regulators of cellular functions, including differentiation and tissue-specific adaptation. The interplay between mitochondria and the nucleus is crucial for coordinating these processes, particularly through the supply of metabolites for epigenetic modifications that facilitate nuclear-mitochondrial interactions. To investigate tissue-specific mitochondrial adaptations at the molecular level, we conducted RNA sequencing data analyses of kidney, heart, brain, and ovary tissues of female buffaloes, focusing on variations in mitochondrial gene expression related to amino acid metabolism. Our analysis identified 82 nuclear-encoded mitochondrial transcripts involved in amino acid metabolism, with significant differential expression patterns across all tissues. Notably, the heart, brain, and kidney-tissues with higher energy demands-exhibited elevated expression levels compared to the ovary. The kidney displayed unique gene expression patterns, characterized by up-regulation of genes involved in glyoxylate metabolism and amino acid catabolism. In contrast, comparative analysis of the heart and kidney versus the brain revealed shared up-regulation of genes associated with fatty acid oxidation. Gene ontology and KEGG pathway analyses confirmed the enrichment of genes in pathways related to amino acid degradation and metabolism. These findings highlight the tissue-specific regulation of mitochondrial gene expression linked to amino acid metabolism, reflecting mitochondrial adaptations to the distinct metabolic and energy requirements of different tissues in buffalo. Importantly, our results underscore the relevance of mitochondrial adaptations not only for livestock health but also for understanding metabolic disorders in humans. By elucidating the molecular mechanisms of mitochondrial function and their tissue-specific variations, this study provides insights that could inform breeding strategies for enhanced livestock productivity and contribute to therapeutic approaches for human metabolic diseases. Thus, our findings illustrate how mitochondria are specialized in a tissue-specific manner to optimize amino acid utilization and maintain cellular homeostasis, with implications for both animal welfare and human health.
Collapse
Affiliation(s)
- E M Sadeesh
- Laboratory of Mitochondrial Biology of Farm Animals, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India.
| | - Madhuri S Lahamge
- Laboratory of Mitochondrial Biology of Farm Animals, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| |
Collapse
|
10
|
Johnson EE, Ervasti JM. Stress exposure in the mdx mouse model of Duchenne muscular dystrophy provokes a widespread metabolic response. FEBS J 2025. [PMID: 39985215 DOI: 10.1111/febs.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/13/2024] [Accepted: 02/07/2025] [Indexed: 02/24/2025]
Abstract
Duchenne muscular dystrophy is a severe neuromuscular wasting disease that is caused by a primary defect in dystrophin protein and involves organism-wide comorbidities such as cardiomyopathy, metabolic and mitochondrial dysfunction, and nonprogressive cognitive impairments. Physiological stress exposure in the mdx mouse model of Duchenne muscular dystrophy results in phenotypic abnormalities that include locomotor inactivity, hypotension, and increased morbidity. Severe and lethal stress susceptibility in mdx mice corresponds to metabolic dysfunction in several coordinated metabolic pathways within dystrophin-deficient skeletal muscle, as well as prolonged elevation in mdx plasma corticosterone levels that extends beyond the wild-type (WT) stress response. Here, we performed a targeted mass spectrometry-based plasma metabolomics screen focused on biological stress pathways in healthy and dystrophin-deficient mdx mice exposed to mild scruff stress. One-third of the stress-relevant metabolites interrogated displayed significant elevation or depletion in mdx plasma after scruff stress and were restored to WT levels by skeletal muscle-specific dystrophin expression. The metabolic pathways of mdx mice altered by scruff stress are associated with regulation of the hypothalamic-pituitary-adrenal axis, locomotor tone, neurocognitive function, redox metabolism, cellular bioenergetics, and protein catabolism. Our data suggest that a mild stress triggers an exaggerated, multi-system metabolic response in mdx mice.
Collapse
Affiliation(s)
- Erynn E Johnson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, USA
| |
Collapse
|
11
|
Liu K, Borreggine R, Gallart-Ayala H, Ivanisevic J, Marques-Vidal P. Serum branched-chain amino acids are mainly associated with body mass index and waist circumference. Nutr Metab Cardiovasc Dis 2025:103880. [PMID: 40087041 DOI: 10.1016/j.numecd.2025.103880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND AND AIMS To assess the associations between serum concentrations of branched-chain amino acids (BCAAs)-valine, leucine, and isoleucine-and different anthropometric markers, including leptin and adiponectin levels, as well as body composition. METHODS AND RESULTS This cross-sectional study used data from the CoLaus|PsyCoLaus and the OsteoLaus studies in Lausanne, Switzerland. Anthropometric markers included the conicity index (CI), body roundness index (BRI), a body shape index (ABSI), body mass index (BMI), and waist circumference, among others. Grip strength was used as a proxy for muscle mass. Bivariate analysis revealed that most anthropometric markers were positively correlated with BCAA, while adiponectin levels was negatively correlated with BCAA. These correlations were generally stronger in males than in females, with the exceptions of CI, ABSI, and adiponectin. After multivariable analysis, weight and BMI showed the strongest association coefficients with BCAA in males, while in females the strongest associations were found for waist circumference and waist-to-height ratio. No significant associations were found between the ABSI and BCAA levels in males, or between grip strength and BCAA levels in females. Stepwise linear regression identified BMI in males, and waist circumference in females as the anthropometric markers most strongly positively associated with BCAA. CONCLUSION We observed a significant difference in the association between BCAA levels and anthropometric markers by sex. BMI displayed the strongest positive association with BCAA levels in males while in females, waist circumference exhibited the strongest association.
Collapse
Affiliation(s)
- Keyuan Liu
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
| | - Rebecca Borreggine
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
12
|
Sato A, Yuyama K, Ichiba Y, Kakizawa Y, Sugiura Y. Branched-chain amino acids and specific phosphatidylinositols are plasma metabolite pairs associated with menstrual pain severity. Sci Rep 2025; 15:3194. [PMID: 39863658 PMCID: PMC11762980 DOI: 10.1038/s41598-025-87415-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
Menstrual pain affects women's quality of life and productivity, yet objective molecular markers for its severity have not been established owing to the variability in blood levels and chemical properties of potential markers such as plasma steroid hormones, lipid mediators, and hydrophilic metabolites. To address this, we conducted a metabolomics study using five analytical methods to identify biomarkers that differentiate menstrual pain severity. This study included 20 women, divided into mild (N = 12) and severe (N = 8) pain groups based on their numerical pain rating scale. We developed pretreatment procedures that allowed all analyses from only 100 µL of finger-prick blood collected across the menstrual cycle. Among the 692 quantified metabolites, branched-chain amino acids and specific phosphatidylinositol (PI), especially PI(36:2), were identified as potential biomarkers. Furthermore, the ratio of PI(36:2) to each BCAA or total BCAA effectively discriminated between the severity levels of menstrual pain. These ratios correlated positively with NPRS, indicating high accuracy in pain assessment. This study highlights the potential of small molecular markers to objectively assess menstrual pain severity, aiding evidence-based support and intervention.
Collapse
Affiliation(s)
- Atsushi Sato
- Research & Development Headquarters, Advanced Analytical Science Research Laboratories, Lion Corporation, Tokyo, Japan.
| | - Kanako Yuyama
- Research & Development Headquarters, Advanced Analytical Science Research Laboratories, Lion Corporation, Tokyo, Japan
| | - Yuko Ichiba
- Research & Development Headquarters, Strategy Management Department, Lion Corporation, Tokyo, Japan
| | - Yasushi Kakizawa
- Research & Development Headquarters, Advanced Analytical Science Research Laboratories, Lion Corporation, Tokyo, Japan
| | - Yuki Sugiura
- Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
- Human Biology Microbiome Quantum Research Center (WPI-Bio2Q), Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
13
|
Brahmachary PP, Erdogan AE, Myers EP, June RK. Metabolomic Profiling and Characterization of a Novel 3D Culture System for Studying Chondrocyte Mechanotransduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.10.598340. [PMID: 38915493 PMCID: PMC11195103 DOI: 10.1101/2024.06.10.598340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Background/Objective Articular chondrocytes synthesize and maintain the avascular and aneural articular cartilage. In vivo these cells are surrounded by a 3D pericellular matrix (PCM) containing predominantly collagen VI. The PCM protects chondrocytes and facilitates mechanotransduction. PCM stiffness is critical in transmitting biomechanical signals to chondrocytes. Various culture systems with different hydrogels are used to encapsulate chondrocytes for 3D culture, but many lack either the PCM or the in vivo stiffness of the cartilage matrix. This study aimed at establishing a culture system to investigate a) if chondrocytes cultured in alginate will develop a PCM and b) study mechanotransduction via metabolic changes induced in 3D agarose-embedded chondrocytes upon mechanical stimulation. Methods We cultured primary human and bovine chondrocytes in monolayers or as alginate encapsulated cells in media containing sodium L-ascorbate. PCM expression was analyzed by immunofluorescence and western blots. We further characterized the response of chondrocytes embedded in physiologically stiff agarose to dynamic compression through metabolomic profiling. Results We found that primary human and bovine chondrocytes, when cultured in alginate beads with addition of sodium L-ascorbate for 7 days, had a pronounced PCM, retained their phenotype, and synthesized both collagens VI and II. This novel culture system enables alginate-encapsulated chondrocytes to develop a robust PCM thereby creating a model system to study mechanotransduction in the presence of an endogenous PCM. We also observed distinct compression-induced changes in metabolomic profiles between the monolayer-agarose and alginate-released agarose-embedded chondrocytes indicating physiological changes in cell metabolism. Conclusion/Significance These data show that 3D preculture of chondrocytes in alginate before encapsulation in physiologically stiff agarose leads to pronounced development of pericellular matrix that is sustained in the presence of ascorbate. This model can be useful in studying the mechanism by which chondrocytes respond to cyclical compression and other types of loading simulating in vivo physiological conditions.
Collapse
Affiliation(s)
- Priyanka P Brahmachary
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT 59717
| | - Ayten E Erdogan
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT 59717
| | - Erik P Myers
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT 59717
| | - Ronald K June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT 59717
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| |
Collapse
|
14
|
Uno K, Uchino T, Suzuki T, Sayama Y, Edo N, Uno-Eder K, Morita K, Ishikawa T, Koizumi M, Honda H, Katagiri H, Tsukamoto K. Rspo3-mediated metabolic liver zonation regulates systemic glucose metabolism and body mass in mice. PLoS Biol 2025; 23:e3002955. [PMID: 39854351 PMCID: PMC11759367 DOI: 10.1371/journal.pbio.3002955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 11/27/2024] [Indexed: 01/26/2025] Open
Abstract
The unique architecture of the liver consists of hepatic lobules, dividing the hepatic features of metabolism into 2 distinct zones, namely the pericentral and periportal zones, the spatial characteristics of which are broadly defined as metabolic zonation. R-spondin3 (Rspo3), a bioactive protein promoting the Wnt signaling pathway, regulates metabolic features especially around hepatic central veins. However, the functional impact of hepatic metabolic zonation, regulated by the Rspo3/Wnt signaling pathway, on whole-body metabolism homeostasis remains poorly understood. In this study, we analyze the local functions of Rspo3 in the liver and the remote actions of hepatic Rspo3 on other organs of the body by using murine models. Rspo3 expression analysis shows that Rspo3 expression patterns are spatiotemporally controlled in the murine liver such that it locates in the pericentral zones and converges after feeding, and the dynamics of these processes are disturbed in obesity. We find that viral-mediated induction of Rspo3 in hepatic tissue of obesity improves insulin resistance and prevents body weight gain by restoring attenuated organ insulin sensitivities, reducing adipose tissue enlargement and reversing overstimulated adaptive thermogenesis. Denervation of the hepatic vagus suppresses these remote effects, derived from hepatic Rspo3 induction, toward adipose tissues and skeletal muscle, suggesting that signals are transduced via the neuronal communication consisting of afferent vagal and efferent sympathetic nerves. Furthermore, the non-neuronal inter-organ communication up-regulating muscle lipid utilization is partially responsible for the ameliorations of both fatty liver development and reduced skeletal muscle quality in obesity. In contrast, hepatic Rspo3 suppression through Cre-LoxP-mediated recombination system exacerbates diabetes due to glucose intolerance and insulin resistance, promotes fatty liver development and decreases skeletal muscle quality, resulting in obesity. Taken together, our study results reveal that modulation of hepatic Rspo3 contributes to maintaining systemic glucose metabolism and body composition via a newly identified inter-organ communication mechanism.
Collapse
Affiliation(s)
- Kenji Uno
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takuya Uchino
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takashi Suzuki
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Yohei Sayama
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Naoki Edo
- Teikyo Academic Research Center, Tokyo, Japan
| | | | - Koji Morita
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Toshio Ishikawa
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Miho Koizumi
- Field of Human Disease Models, Tokyo Women’s Medical University, Tokyo, Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Tokyo Women’s Medical University, Tokyo, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhisa Tsukamoto
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Wunderle C, Ciobanu C, Ritz J, Tribolet P, Neyer P, Bernasconi L, Stanga Z, Mueller B, Schuetz P. Association of leucine and other branched chain amino acids with clinical outcomes in malnourished inpatients: a secondary analysis of the randomized clinical trial EFFORT. Eur J Clin Nutr 2025; 79:42-49. [PMID: 39245679 DOI: 10.1038/s41430-024-01507-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND The essential branched-chain amino acids leucine, isoleucine and valine are considered anabolic and stimulate protein synthesis in the muscles as well in the liver. They also promote muscle recovery and contribute to glucose homeostasis. Recent studies in critically ill patients have demonstrated that depletion of plasma leucine is associated with increased mortality, but data in the non-critical care setting is lacking. METHODS This secondary analysis of the randomized controlled Effect of early nutritional support on Frailty, Functional Outcomes, and Recovery of malnourished medical inpatients Trial (EFFORT), investigated the impact of leucine, isoleucine, and valine metabolism on clinical outcomes. The primary endpoint was 180-day all-cause mortality. RESULTS Among 238 polymorbid patients with available metabolite measurements, low serum leucin levels were associated with a doubled risk of 180-day all-cause mortality in a fully adjusted regression model (adjusted HR 2.20 [95% CI 1.46-3.30], p < 0.001). There was also an association with mortality for isoleucine (1.56 [95% CI 1.03-2.35], p = 0.035) and valine (1.69 [95% CI 1.13-2.53], p = 0.011). When comparing effects of nutritional support on mortality in patients with high and low levels of leucine, there was no evidence of significant differences in effectiveness of the intervention. The same was true for isoleucine and valine. CONCLUSION Our data suggest that depletion of leucine, isoleucine, and valine among malnourished polymorbid patients is associated with increases in long-term mortality. However, patients with low metabolite levels did not show a pronounced benefit from nutritional support. Further research should focus on the clinical effects of nutritional support in patients with depleted stores of essential branched-chain amino acids. CLINICAL TRIAL REGISTRATION clinicaltrials.gov as NCT02517476 (registered 7 August 2015).
Collapse
Affiliation(s)
- Carla Wunderle
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
| | - Claudia Ciobanu
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Medical Faculty of the University of Basel, Basel, Switzerland
| | - Jacqueline Ritz
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Medical Faculty of the University of Basel, Basel, Switzerland
| | - Pascal Tribolet
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Department of Health Professions, Bern University of Applied Sciences, Bern, Switzerland
- Department of Nutritional Sciences and Research Platform Active Ageing, University of Vienna, Vienna, Austria
| | - Peter Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Zeno Stanga
- Division of Diabetes, Endocrinology, Nutritional Medicine, and Metabolism, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Beat Mueller
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Medical Faculty of the University of Basel, Basel, Switzerland
| | - Philipp Schuetz
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland.
- Medical Faculty of the University of Basel, Basel, Switzerland.
| |
Collapse
|
16
|
Rodrigo‐Carbó C, Madinaveitia‐Nisarre L, Pérez‐Calahorra S, Gracia‐Rubio I, Cebollada A, Galindo‐Lalana C, Mateo‐Gallego R, Lamiquiz‐Moneo I. Low-calorie, high-protein diets, regardless of protein source, improve glucose metabolism and cardiometabolic profiles in subjects with prediabetes or type 2 diabetes and overweight or obesity. Diabetes Obes Metab 2025; 27:268-279. [PMID: 39420528 PMCID: PMC11618321 DOI: 10.1111/dom.16013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Abstract
AIM The aim was to study the effect of two low-calorie, high-protein (HP) diets, with most of the protein coming from animal or plant sources, on glycaemic and other cardiometabolic outcomes in subjects with overweight or obesity and glucose metabolism disorders. MATERIALS AND METHODS A total of 117 participants aged >18 years with body mass index over 27.5 kg/m2 and prediabetes or type 2 diabetes mellitus (T2DM) were randomized to one of two HP low-calorie diets (35% of total calories from protein), in which 75% of the protein was from either plant-based sources (HPP) or animal sources (HPA). For both diets, 30% and 35% of the total calories were from fat and carbohydrates, respectively. The dietary intervention lasted 6 months. RESULTS Both diets improved body composition to a similar extent, including weight loss (-8.05 ± 5.12 kg for the HPA diet and -7.70 ± 5.47 kg for the HPP diet at 6 months) and fat mass, mainly visceral fat. Both diets had a similar beneficial effect on glucose metabolism, including fasting glucose, insulin, homeostasis model assessment of insulin resistance index and glycated haemoglobin. Other biochemical parameters, including lipid profiles, liver enzymes, adipokines and inflammatory biomarkers, similarly improved in both groups. Fasting incretins, mainly glucagon-like peptide 1, decreased significantly in both groups, and this effect correlated with weight loss. CONCLUSIONS Low-calorie HP diets improved body composition, glucose metabolism and other cardiometabolic outcomes, regardless of protein source (either animal or plant sources), in outpatients with prediabetes or T2DM. CLINICAL TRIAL REGISTRATION The clinical trial was registered in ClinicalTrials.gov (identifier: NCT05456347) https://clinicaltrials.gov/study/NCT05456347?term=NCT05456347&rank=1.
Collapse
Affiliation(s)
- Carmen Rodrigo‐Carbó
- Miguel Servet University Hospital, Aragon Health Research Institute, CIBERCVZaragozaSpain
- Departament of Physiatry and Nursing, Faculty of Health and Sport ScienceUniversity of ZaragozaHuescaSpain
| | | | - Sofía Pérez‐Calahorra
- Departament of Physiatry and Nursing, Faculty of Health ScienceUniversity of ZaragozaZaragozaSpain
| | - Irene Gracia‐Rubio
- Miguel Servet University Hospital, Aragon Health Research Institute, CIBERCVZaragozaSpain
- Department of Human Anatomy and Histology, Faculty of MedicineUniversity of ZaragozaZaragozaSpain
| | | | - Carlos Galindo‐Lalana
- Miguel Servet University Hospital, Aragon Health Research Institute, CIBERCVZaragozaSpain
| | - Rocío Mateo‐Gallego
- Miguel Servet University Hospital, Aragon Health Research Institute, CIBERCVZaragozaSpain
- Departament of Physiatry and Nursing, Faculty of Health and Sport ScienceUniversity of ZaragozaHuescaSpain
| | - Itziar Lamiquiz‐Moneo
- Miguel Servet University Hospital, Aragon Health Research Institute, CIBERCVZaragozaSpain
- Department of Human Anatomy and Histology, Faculty of MedicineUniversity of ZaragozaZaragozaSpain
| |
Collapse
|
17
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
18
|
da Cruz-Filho J, Costa DM, Santos TO, da Silva RP, Anjos-Santos HC, Marciano NJDS, Rodríguez-Gúzman R, Henrique-Santos AB, Melo JEC, Badauê-Passos D, Murphy D, Mecawi AS, Lustrino D. Water deprivation induces a systemic procatabolic state that differentially affects oxidative and glycolytic skeletal muscles in male mice. Am J Physiol Regul Integr Comp Physiol 2025; 328:R21-R33. [PMID: 39466171 DOI: 10.1152/ajpregu.00187.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024]
Abstract
Dehydration, characterized by the loss of total body water and/or electrolytes due to diseases or inadequate fluid intake, is prevalent globally but often underestimated. Its contribution to long-term chronic diseases and sarcopenia is recognized, yet the mechanisms involved in systemic and muscle protein metabolism during dehydration remain unclear. This study investigated metabolic adaptations in a 36-h water deprivation (WD) model of mice. Male C57BL/6 mice underwent 36-h WD or pair-feeding at rest, with assessments of motor skills along with biochemical and metabolic parameters. Dehydration was confirmed by hypernatremia, body mass loss, hyporexia, and increased activity of vasopressinergic and oxytocinergic neurons compared with controls. These results were associated with liver mass loss, decreased glycemia, and increased cholesterolemia. In addition, increased V̇o2 and a decreased respiratory exchange ratio indicated reduced carbohydrate consumption and potentially increased protein use during dehydration. Thus, skeletal muscle protein metabolism was evaluated due to its high protein content. In the oxidative muscles of the WD group, total and proteasomal proteolysis increased, which was associated with decreased Akt-mediated intracellular signaling. Interestingly, there was an increase in fiber cross-sectional area, likely due to higher muscle water content caused by increased intracellular osmolality induced by protein catabolism products. Conversely, no changes were observed in protein turnover or water content in glycolytic muscles. These findings suggest that short-term WD imposes a procatabolic state, depleting protein content in skeletal muscle. However, skeletal muscle may respond differently to dehydration based on its phenotype and might adapt for a limited time.NEW & NOTEWORTHY This study investigated the effects of WD on mouse homeostasis, focusing on energy substrates and skeletal muscle protein metabolism. Our findings revealed a shift toward reduced dependence on carbohydrate degradation and increased reliance on lipid oxidation, or even protein oxidation, as energy sources, since we observed increased proteolysis in one muscle phenotype. Despite body mass loss, soleus and EDL muscle masses were differently affected. These results indicate the procatabolic potential of short-term WD in mice.
Collapse
Affiliation(s)
- João da Cruz-Filho
- Laboratory of Basic and Behavioural Neuroendocrinology (LANBAC), Department of Physiology, Centre for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil
- Graduate Program in Physiological Sciences (PROCFIS), Federal University of Sergipe, São Cristóvão, Brazil
| | - Daniely Messias Costa
- Laboratory of Basic and Behavioural Neuroendocrinology (LANBAC), Department of Physiology, Centre for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil
- Graduate Program in Physiological Sciences (PROCFIS), Federal University of Sergipe, São Cristóvão, Brazil
| | - Tatiane Oliveira Santos
- Laboratory of Basic and Behavioural Neuroendocrinology (LANBAC), Department of Physiology, Centre for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil
- Graduate Program in Physiological Sciences (PROCFIS), Federal University of Sergipe, São Cristóvão, Brazil
| | - Raquel Prado da Silva
- Laboratory of Basic and Behavioural Neuroendocrinology (LANBAC), Department of Physiology, Centre for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil
- Graduate Program in Physiological Sciences (PROCFIS), Federal University of Sergipe, São Cristóvão, Brazil
| | - Hevely Catharine Anjos-Santos
- Laboratory of Basic and Behavioural Neuroendocrinology (LANBAC), Department of Physiology, Centre for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil
- Graduate Program in Physiological Sciences (PROCFIS), Federal University of Sergipe, São Cristóvão, Brazil
| | - Naima Jamile Dos Santos Marciano
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, São Paulo School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Roger Rodríguez-Gúzman
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, São Paulo School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ana Beatriz Henrique-Santos
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, São Paulo School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - João Eduardo Conceição Melo
- Laboratory of Basic and Behavioural Neuroendocrinology (LANBAC), Department of Physiology, Centre for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil
- Graduate Program in Physiological Sciences (PROCFIS), Federal University of Sergipe, São Cristóvão, Brazil
| | - Daniel Badauê-Passos
- Laboratory of Basic and Behavioural Neuroendocrinology (LANBAC), Department of Physiology, Centre for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - André Souza Mecawi
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, São Paulo School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Danilo Lustrino
- Laboratory of Basic and Behavioural Neuroendocrinology (LANBAC), Department of Physiology, Centre for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil
- Graduate Program in Physiological Sciences (PROCFIS), Federal University of Sergipe, São Cristóvão, Brazil
| |
Collapse
|
19
|
Abuelazm M, Fares A, Elhady MM, Amin AM, Khan U, Gowaily I, Jaber F. Branched-Chain Amino Acid Supplements for Sarcopenia in Liver Cirrhosis: A Systematic Review and Meta-analysis. J Clin Exp Hepatol 2025; 15:102417. [PMID: 39834601 PMCID: PMC11742309 DOI: 10.1016/j.jceh.2024.102417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/23/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Sarcopenia, a key aspect of malnutrition in liver cirrhosis (LC), affects 30-70% of LC patients. Given the inconsistent results from RCTs on branched-chain amino acids (BCAAs) for treating sarcopenia in LC, we conducted a systematic review and meta-analysis to assess the efficacy and safety of BCAAs for sarcopenia management in LC patients. METHODS A systematic review and meta-analysis synthesizing evidence from RCTs obtained from PubMed, Embase, Cochrane, Scopus, and Web of Science from inception to April 2024. We used the fixed-effects model to report dichotomous outcomes using risk ratio (RR) and continuous outcomes using mean difference (MD), with a 95% confidence interval (CI). PROSPERO ID: CRD42024542761. RESULTS Five RCTs with 434 patients were included. BCAAs were significantly associated with decreased liver frailty index change (MD: -0.14 with 95% CI [-0.28, -0.01], P = 0.03). However, there was no significant difference between BCAAs and the control group regarding hand grip strength change (MD: 0.98 with 95% CI [-0.45, 2.41], P = 0.18). Also, BCAAs were associated with increased body mass index (BMI) change (MD: 0.99 with 95% CI [0.16, 1.82], P = 0.02) and increased QoL (standardized mean difference : 0.27 with 95% CI [0.03, 0.52], P = 0.03). However, there was no significant difference between BCAAs and the control group in model for end-stage liver disease (MELD) score change (MD: 0.65 with 95% CI [-1.20, 2.50], P = 0.49), skeletal muscle index change (MD: 0.21 with 95% CI [-0.23, 0.65], P = 0.35), and gait speed change (MD: 0.10 with 95% CI [-0.15, 0.34], P = 0.43). CONCLUSION BCAA supplementation in cirrhotic patients with sarcopenia reduced the liver frailty index, increased BMI and QoL, but did not affect handgrip strength, skeletal muscle index, gait speed, or MELD score. Outcome heterogeneity and study bias were noted, highlighting the need for further RCTs to confirm these results.
Collapse
Affiliation(s)
| | - Ahmed Fares
- Internal Medicine Department, Tufts Medical Center, Boston, USA
| | | | | | - Ubaid Khan
- Department of Medicine, University of Maryland School of Medicine, USA
| | | | - Fouad Jaber
- Department of Gastroenterology and hepatology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
20
|
Tan EY, Muthiah MD, Sanyal AJ. Metabolomics at the cutting edge of risk prediction of MASLD. Cell Rep Med 2024; 5:101853. [PMID: 39657668 PMCID: PMC11722125 DOI: 10.1016/j.xcrm.2024.101853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/12/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major public health threat globally. Management of patients afflicted with MASLD and research in this domain are limited by the lack of robust well-established non-invasive biomarkers for diagnosis, prognostication, and monitoring. The circulating metabolome reflects both the systemic metabo-inflammatory milieu and changes in the liver in affected individuals. In this review we summarize the available literature on changes in the different components of the metabolome in MASLD with a focus on changes that are linked to the presence of underlying steatohepatitis, severity of disease activity, and fibrosis stage. We further summarize the existing literature around biomarker panels that are derived from interrogation of the metabolome. Their relevance to disease biology and utility in practice are also discussed. We further highlight potential direction for future studies particularly to ensure they are fit for purpose and suitable for widespread use.
Collapse
Affiliation(s)
- En Ying Tan
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore.
| | - Mark D Muthiah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
21
|
Sato Y, Hishiki T, Masugi Y, Florence L, Yu YM. Vitamin D administration increases serum alanine concentrations in thermally injured mice. Biochem Biophys Res Commun 2024; 736:150505. [PMID: 39128265 DOI: 10.1016/j.bbrc.2024.150505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Thermal or burn injury results in profound metabolic changes in the body. This can contribute to muscle atrophy, bone loss, as well as suppression of the immune system. While the mechanisms that underlie this hypermetabolic response remain unclear, patients with burn injury often have low circulating levels of vitamin D. Vitamin D has been shown to regulate bone formation as well as regulate muscle function. We sought to clarify the effects of vitamin D administration on skeletal muscle function following thermal injury using a mouse model. We found that thermal injury resulted in decreased vitamin D levels as well as decreased bone mineral density. Branched chain amino acid (BCAA)s levels were also significantly enhanced in the serum following burn injury. Vitamin D administration reversed the decrease in bone marrow-derived mesenchymal stem cell (BM-MSC)s observed post burn injury. Interestingly, vitamin D administration also resulted in increased tricarboxylic acid cycle (TCA) cycle metabolites in muscle which was decreased after burn conditions, enhanced the supply of alanine and glutamine in the blood which could contribute to gluconeogenesis and wound healing. Therefore, vitamin D supplementation after burn injury may have effects not only in bone metabolism, but may affect substrate metabolism in other organs/tissues.
Collapse
Affiliation(s)
- Yukio Sato
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo, Japan; Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Hospital for Children in Boston, Boston, MA, USA.
| | - Takako Hishiki
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Masugi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Lin Florence
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Hospital for Children in Boston, Boston, MA, USA
| | - Yong-Ming Yu
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Hospital for Children in Boston, Boston, MA, USA
| |
Collapse
|
22
|
Marsh NM, MacEwen MJS, Chea J, Kenerson HL, Kwong AA, Locke TM, Miralles FJ, Sapre T, Gozali N, Hart ML, Bammler TK, MacDonald JW, Sullivan LB, Atilla-Gokcumen GE, Ong SE, Scott JD, Yeung RS, Sancak Y. Mitochondrial Calcium Signaling Regulates Branched-Chain Amino Acid Catabolism in Fibrolamellar Carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.596106. [PMID: 38853984 PMCID: PMC11160645 DOI: 10.1101/2024.05.27.596106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Metabolic adaptations in response to changes in energy supply and demand are essential for survival. The mitochondrial calcium uniporter plays a key role in coordinating metabolic homeostasis by regulating TCA cycle activation, mitochondrial fatty acid oxidation, and cellular calcium signaling. However, a comprehensive analysis of uniporter-regulated mitochondrial pathways has remained unexplored. Here, we investigate metabolic consequences of uniporter loss- and gain-of-function using uniporter knockout cells and the liver cancer fibrolamellar carcinoma (FLC), which we demonstrate to have elevated mitochondrial calcium levels. Our results reveal that branched-chain amino acid (BCAA) catabolism and the urea cycle are uniporter-regulated metabolic pathways. Reduced uniporter function boosts expression of BCAA catabolism genes, and the urea cycle enzyme ornithine transcarbamylase (OTC). In contrast, high uniporter activity in FLC suppresses their expression. This suppression is mediated by reduced expression of the transcription factor KLF15, a master regulator of liver metabolism. Thus, uniporter responsive calcium signaling plays a central role in FLC-associated metabolic changes, including hyperammonemia. Our study identifies an important role for mitochondrial calcium signaling in metabolic adaptation through transcriptional regulation of metabolism and elucidates its importance for BCAA and ammonia metabolism in FLC.
Collapse
Affiliation(s)
- Nicole M Marsh
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Melissa J S MacEwen
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Jane Chea
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Heidi L Kenerson
- Department of Surgery, University of Washington Medical Center, Seattle, WA, United States
| | - Albert A Kwong
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Timothy M Locke
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | | | - Tanmay Sapre
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Natasha Gozali
- Department of Chemistry, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Madeleine L Hart
- Human Biology Division, Fred Hutchinson Cancer Center, WA, Seattle, United States
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Lucas B Sullivan
- Human Biology Division, Fred Hutchinson Cancer Center, WA, Seattle, United States
| | - G Ekin Atilla-Gokcumen
- Department of Chemistry, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Shao-En Ong
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - John D Scott
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Raymond S Yeung
- Department of Surgery, University of Washington Medical Center, Seattle, WA, United States
| | - Yasemin Sancak
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
23
|
Li Z, Chen S, Wu X, Liu F, Zhu J, Chen J, Lu X, Chi R. Research advances in branched-chain amino acid metabolism in tumors. Mol Cell Biochem 2024. [DOI: 10.1007/s11010-024-05163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/10/2024] [Indexed: 01/06/2025]
|
24
|
Karvinen S, Korhonen TMK, Kiviö R, Lensu S, Gajera B, Britton SL, Koch LG, Nieminen AI, Kainulainen H. Branched-chain amino acid supplementation and voluntary running have distinct effects on the serum metabolome of rats with high or low intrinsic aerobic capacity. Front Nutr 2024; 11:1450386. [PMID: 39628463 PMCID: PMC11611553 DOI: 10.3389/fnut.2024.1450386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/10/2024] [Indexed: 12/06/2024] Open
Abstract
Introduction A growing body of literature associates branched-chain amino acid (BCAA) catabolism to increased fatty acid oxidation and better metabolic health. Hence, BCAA-rich diets may improve body composition and muscle protein synthesis. However, the role of individual characteristics such as a low aerobic fitness, a well-established risk factor for cardio-metabolic diseases, has not been studied. Methods This study examined 64 female rats from the high-capacity runner (HCR) and low-capacity runner (LCR) rat model. Rats from each line (HCR or LCR) were divided into four groups; differing from diet (CTRL or BCAA) and from the opportunity to voluntarily run on a running wheel (NONRUNNER or RUNNER). Groups were matched for body mass and maximal running capacity within each line. We measured maximal running capacity and metabolism before and after the intervention of diet and voluntary running activity. After the end of the experiment, serum samples were collected for metabolome analysis. Results We are the first to show that BCAA supplementation has a more pronounced impact on LCRs compared to HCRs. Specifically, in LCR rats, BCAA supplementation led to reduced daily voluntary running distance and an enrichment of serine metabolism in the serum metabolome. While voluntary running increased food intake and energy expenditure, its effects on the serum metabolome were minimal in HCRs. Conclusion The present research highlights the benefit achieved by combining BCAA supplementation with running activity, especially in the LCR line. Importantly, our results underscore the interconnected role of BCAAs and fatty acid metabolism in promoting overall metabolic health.
Collapse
Affiliation(s)
- Sira Karvinen
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Tia-Marje K. Korhonen
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Ronja Kiviö
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Sanna Lensu
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
- Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| | - Bharat Gajera
- Metabolomics Unit, Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Steven L. Britton
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Anaesthesiology, University of Michigan, Ann Arbor, MI, United States
| | - Lauren G. Koch
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, United States
| | - Anni I. Nieminen
- Metabolomics Unit, Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Heikki Kainulainen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
25
|
Mironchuk O, Chang AL, Rahmani F, Portell K, Nunez E, Nigogosyan Z, Ma D, Popuri K, Chow VTY, Beg MF, Luo J, Ippolito JE. Volumetric body composition analysis of the Cancer Genome Atlas reveals novel body composition traits and molecular markers Associated with Renal Carcinoma outcomes. Sci Rep 2024; 14:27022. [PMID: 39505904 PMCID: PMC11541764 DOI: 10.1038/s41598-024-76280-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024] Open
Abstract
Clinically, the body mass index remains the most frequently used metric of overall obesity, although it is flawed by its inability to account for different adipose (i.e., visceral, subcutaneous, and inter/intramuscular) compartments, as well as muscle mass. Numerous prior studies have demonstrated linkages between specific adipose or muscle compartments to outcomes of multiple diseases. Although there are no universally accepted standards for body composition measurement, many studies use a single slice at the L3 vertebral level. In this study, we use computed tomography (CT) studies from patients in The Cancer Genome Atlas (TCGA) to compare current L3-based techniques with volumetric techniques, demonstrating potential limitations with level-based approaches for assessing outcomes. In addition, we identify gene expression signatures in normal kidney that correlate with fat and muscle body composition traits that can be used to predict sex-specific outcomes in renal cell carcinoma.
Collapse
Affiliation(s)
| | - Andrew L Chang
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA
| | - Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA
| | - Kaitlyn Portell
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA
| | - Elena Nunez
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA
| | - Zack Nigogosyan
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA
| | - Da Ma
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | | | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Siteman Cancer Center Biostatistics and Qualitative Research Shared Resource, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
26
|
Axelrod CL, Hari A, Dantas WS, Kashyap SR, Schauer PR, Kirwan JP. Metabolomic Fingerprints of Medical Therapy Versus Bariatric Surgery in Patients With Obesity and Type 2 Diabetes: The STAMPEDE Trial. Diabetes Care 2024; 47:2024-2032. [PMID: 39311919 PMCID: PMC11502526 DOI: 10.2337/dc24-0859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVE Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) are effective procedures to treat and manage type 2 diabetes (T2D). However, the underlying metabolic adaptations that mediate improvements in glucose homeostasis remain largely elusive. The purpose of this study was to identify metabolic signatures associated with biochemical resolution of T2D after medical therapy (MT) or bariatric surgery. RESEARCH DESIGN AND METHODS Plasma samples from 90 patients (age 49.9 ± 7.6 years; 57.7% female) randomly assigned to MT (n = 30), RYGB (n = 30), or SG (n = 30) were retrospectively subjected to untargeted metabolomic analysis using ultra performance liquid chromatography with tandem mass spectrometry at baseline and 24 months of treatment. Phenotypic importance was determined by supervised machine learning. Associations between change in glucose homeostasis and circulating metabolites were assessed using a linear mixed effects model. RESULTS The circulating metabolome was dramatically remodeled after SG and RYGB, with largely overlapping signatures after MT. Compared with MT, SG and RYGB profoundly enhanced the concentration of metabolites associated with lipid and amino acid signaling, while limiting xenobiotic metabolites, a function of decreased medication use. Random forest analysis revealed 2-hydroxydecanoate as having selective importance to RYGB and as the most distinguishing feature between MT, SG, and RYGB. To this end, change in 2-hydroxydecanoate correlated with reductions in fasting glucose after RYGB but not SG or MT. CONCLUSIONS We identified a novel metabolomic fingerprint characterizing the longer-term adaptations to MT, RYGB, and SG. Notably, the metabolomic profiles of RYGB and SG procedures were distinct, indicating equivalent weight loss may be achieved by divergent effects on metabolism.
Collapse
Affiliation(s)
- Christopher L. Axelrod
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Adithya Hari
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Wagner S. Dantas
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | | | - Philip R. Schauer
- Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH
- Clinical Metabolic Surgery Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - John P. Kirwan
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
27
|
Chang YC, Chen YC, Chan YC, Liu C, Chang SJ. Oligonol ®, an Oligomerized Polyphenol from Litchi chinensis, Enhances Branched-Chain Amino Acid Transportation and Catabolism to Alleviate Sarcopenia. Int J Mol Sci 2024; 25:11549. [PMID: 39519101 PMCID: PMC11546093 DOI: 10.3390/ijms252111549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Branched-chain amino acids (BCAAs) are essential for muscle protein synthesis and are widely acknowledged for mitigating sarcopenia. Oligonol® (Olg), a low-molecular-weight polyphenol from Litchi chinensis, has also been found to attenuate sarcopenia by improving mitochondrial quality and positive protein turnover. This study aims to investigate the effect of Olg on BCAA-stimulated protein synthesis in sarcopenia. In sarcopenic C57BL/6 mice and senescence-accelerated mouse-prone 8 (SAMP8) mice, BCAAs were significantly decreased in skeletal muscle but increased in blood serum. Furthermore, the expressions of membrane L-type amino acid transporter 1 (LAT1) and branched-chain amino acid transaminase 2 (BCAT2) in skeletal muscle were lower in aged mice than in young mice. The administration of Olg for 8 weeks significantly increased the expressions of membrane LAT1 and BCAT2 in the skeletal muscle when compared with non-treated SAMP8 mice. We further found that BCAA deprivation via LAT1-siRNA in C2C12 myotubes inhibited the signaling of protein synthesis and facilitated ubiquitination degradation of BCAT2. In C2C12 cells mimicking sarcopenia, Olg combined with BCAA supplementation enhanced mTOR/p70S6K activity more than BCAA alone. However, blocked LAT1 by JPH203 reversed the synergistic effect of the combination of Olg and BCAAs. Taken together, changes in LAT1 and BCAT2 during aging profoundly alter BCAA availability and nutrient signaling in aged mice. Olg increases BCAA-stimulated protein synthesis via modulating BCAA transportation and BCAA catabolism. Combining Olg and BCAAs may be a useful nutritional strategy for alleviating sarcopenia.
Collapse
Affiliation(s)
- Yun-Ching Chang
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan; (Y.-C.C.)
| | - Yu-Chi Chen
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan; (Y.-C.C.)
- Department of Urology, E-Da Cancer Hospital, I-Shou University, Kaohsiung 82445, Taiwan
| | - Yin-Ching Chan
- Department of Food and Nutrition, Providence University, Taichung 43330, Taiwan;
| | - Cheng Liu
- Department of Physical Therapy, Shu-Zen Junior College of Medicine and Management, Kaohsiung 82144, Taiwan
| | - Sue-Joan Chang
- Department of Life Sciences, National Cheng Kung University, Tainan 701, Taiwan
- Marine Biology and Cetacean Research Center, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
28
|
Sink WJ, Fling R, Yilmaz A, Nault R, Goniwiecha D, Harkema JR, Graham SF, Zacharewski T. 2,3,7,8-Tetrachlorodibenzo- p-dioxin (TCDD) elicited dose-dependent shifts in the murine urinary metabolome associated with hepatic AHR-mediated differential gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619714. [PMID: 39484576 PMCID: PMC11526911 DOI: 10.1101/2024.10.22.619714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Epidemiological evidence suggests an association between dioxin and dioxin-like compound (DLC) exposure and human liver disease. The prototypical DLC, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), has been shown to induce the progression of reversible hepatic steatosis to steatohepatitis with periportal fibrosis and biliary hyperplasia in mice. Although the effects of TCDD toxicity are mediated by aryl hydrocarbon receptor (AHR) activation, the underlying mechanisms of TCDD-induced hepatotoxicity are unresolved. In the present study, male C57BL/6NCrl mice were gavaged every 4 days for 28 days with 0.03 - 30 μg/kg TCDD and evaluated for liver histopathology and gene expression as well as complementary 1-dimensional proton magnetic resonance (1D- 1H NMR) urinary metabolic profiling. Urinary trimethylamine (TMA), trimethylamine N-oxide (TMAO), and 1-methylnicotinamide (1MN) levels were altered by TCDD at doses ≤ 3 μg/kg; other urinary metabolites, like glycolate, urocanate, and 3-hydroxyisovalerate, were only altered at doses that induced moderate to severe steatohepatitis. Bulk liver RNA-seq data suggested altered urinary metabolites correlated with hepatic differential gene expression corresponding to specific metabolic pathways. In addition to evaluating whether altered urinary metabolites were liver-dependent, published single-nuclear RNA-seq (snRNA-seq), AHR ChIP-seq, and AHR knockout gene expression datasets provide further support for hepatic cell-type and AHR-regulated dependency, respectively. Overall, TCDD-induced liver effects were preceded by and occurred with changes in urinary metabolite levels due to AHR-mediated changes in hepatic gene expression.
Collapse
Affiliation(s)
- Warren J Sink
- Michigan State University, Department of Biochemistry and Molecular Biology, East Lansing, MI 48823, USA
- Michigan State University, Institute for Integrative Toxicology, East Lansing, MI 48824, USA
| | - Russell Fling
- Michigan State University, Department of Biochemistry and Molecular Biology, East Lansing, MI 48823, USA
- Michigan State University, Institute for Integrative Toxicology, East Lansing, MI 48824, USA
| | - Ali Yilmaz
- Corewell Health Research Institute, Royal Oak, MI 48073, USA
| | - Rance Nault
- Michigan State University, Department of Pharmacology and Toxicology, East Lansing, MI 48824, USA
| | - Delanie Goniwiecha
- Middlebury College, Neuroscience Faculty, 14 Old Chapel Rd, Middlebury, VT 05753, USA
| | - Jack R Harkema
- Michigan State University, Pathobiology & Diagnostic Investigation, East Lansing, MI, United States of America
| | - Stewart F Graham
- Corewell Health Research Institute, Royal Oak, MI 48073, USA
- Oakland University-William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Timothy Zacharewski
- Michigan State University, Department of Biochemistry and Molecular Biology, East Lansing, MI 48823, USA
- Michigan State University, Institute for Integrative Toxicology, East Lansing, MI 48824, USA
| |
Collapse
|
29
|
Mann G, Adegoke OAJ. Elevated BCAA catabolism reverses the effect of branched-chain ketoacids on glucose transport in mTORC1-dependent manner in L6 myotubes. J Nutr Sci 2024; 13:e66. [PMID: 39464407 PMCID: PMC11503859 DOI: 10.1017/jns.2024.66] [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: 04/24/2024] [Revised: 08/13/2024] [Accepted: 08/30/2024] [Indexed: 10/29/2024] Open
Abstract
Plasma levels of branched-chain amino acids (BCAA) and their metabolites, branched-chain ketoacids (BCKA), are increased in insulin resistance. We previously showed that ketoisocaproic acid (KIC) suppressed insulin-stimulated glucose transport in L6 myotubes, especially in myotubes depleted of branched-chain ketoacid dehydrogenase (BCKD), the enzyme that decarboxylates BCKA. This suggests that upregulating BCKD activity might improve insulin sensitivity. We hypothesised that increasing BCAA catabolism would upregulate insulin-stimulated glucose transport and attenuate insulin resistance induced by BCKA. L6 myotubes were either depleted of BCKD kinase (BDK), the enzyme that inhibits BCKD activity, or treated with BT2, a BDK inhibitor. Myotubes were then treated with KIC (200 μM), leucine (150 μM), BCKA (200 μM), or BCAA (400 μM) and then treated with or without insulin (100 nM). BDK depletion/inhibition rescued the suppression of insulin-stimulated glucose transport by KIC/BCKA. This was consistent with the attenuation of IRS-1 (Ser612) and S6K1 (Thr389) phosphorylation but there was no effect on Akt (Ser473) phosphorylation. The effect of leucine or BCAA on these measures was not as pronounced and BT2 did not influence the effect. Induction of the mTORC1/IRS-1 (Ser612) axis abolished the attenuating effect of BT2 treatment on glucose transport in cells treated with KIC. Surprisingly, rapamycin co-treatment with BT2 and KIC further reduced glucose transport. Our data suggests that the suppression of insulin-stimulated glucose transport by KIC/BCKA in muscle is mediated by mTORC1/S6K1 signalling. This was attenuated by upregulating BCAA catabolic flux. Thus, interventions targeting BCAA metabolism may provide benefits against insulin resistance and its sequelae.
Collapse
Affiliation(s)
- Gagandeep Mann
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Olasunkanmi A. John Adegoke
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|
30
|
Muscella A, Felline M, Marsigliante S. Sex-Based Effects of Branched-Chain Amino Acids on Strength Training Performance and Body Composition. Sports (Basel) 2024; 12:275. [PMID: 39453241 PMCID: PMC11510782 DOI: 10.3390/sports12100275] [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: 08/08/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Branched-chain amino acids (BCAAs) are widely studied for their effects on muscle recovery and performance. AIMS This study examined the effects of BCAA supplementation on anthropometric data, physical performance, delayed onset muscle soreness (DOMS), and fatigue in recreational weightlifters. METHODS The trial involved 100 participants (50 men and 50 women), randomized into BCAA and placebo groups. Subjects in the BCAA group took five daily capsules of 500 mg L-leucine, 250 mg L-isoleucine, and 250 mg L-valine for six months. A two-way ANOVA was used to analyze the main and interaction effects of sex and treatment. RESULTS Notable findings include significant improvements in muscle recovery, as indicated by reduced DOMS, particularly in women who showed a decrement of 18.1 ± 9.4 mm compared to 0.8 ± 1.2 mm in the placebo group of a horizontal 100 mm line. Fatigue perception was also significantly lower in the BCAA group, with women reporting a greater decrease (2.6 ± 1.5 scores) compared to the placebo group (0.6 ± 0.7 scores). Strength gains were prominent, especially in men, with a 10% increase in bench press maximum observed in the BCAA group. The interaction between sex and treatment was significant, suggesting sex-specific responses to BCAA supplementation. CONCLUSIONS These results underscore the effectiveness of BCAA supplementation in enhancing muscle recovery, reducing fatigue, and improving strength. This study also highlights sex-specific responses, with women benefiting more in terms of DOMS and fatigue reduction, while men experienced greater strength gains, suggesting a need for tailored supplementation strategies.
Collapse
Affiliation(s)
- Antonella Muscella
- Department of Biological and Environmental Science and Technologies (DiSTeBA), University of Salento, 73100 Lecce, Italy (S.M.)
| | | | | |
Collapse
|
31
|
Spears M, Cooper G, Sather B, Bailey M, Boles JA, Bothner B, Miles MP. Comparative Impact of Organic Grass-Fed and Conventional Cattle-Feeding Systems on Beef and Human Postprandial Metabolomics-A Randomized Clinical Trial. Metabolites 2024; 14:533. [PMID: 39452914 PMCID: PMC11509860 DOI: 10.3390/metabo14100533] [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: 08/31/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES Cattle-feeding systems may have health implications for consumers of beef products. Organic grass-fed (GRA) and conventional (CON) cattle-feeding systems may result in beef products with differing metabolite profiles and therefore could impact the postprandial metabolomic response of consumers. This study aims to measure whole beef metabolomics and postprandial metabolomic response of consumers between GRA and CON beef to elucidate potential health implications. METHODS This study followed a randomized double-blind crossover design with healthy male and female subjects (n = 10). Plasma samples were taken at fasting (0) and postprandially for four hours after consumption of a steak from each condition. Untargeted metabolomic analysis of whole beef and human plasma samples used LC/MS. Multivariate and pathway enrichment analysis in MetaboAnalyst was used to investigate metabolite and biochemical pathways that distinguished CON and GRA. RESULTS Cattle-feeding systems impacted both postprandial and whole beef steak metabolomic profiles. Metabolites that contributed to this variation included carnitine species (Proionylcarnitine), fatty acids, amino acids (L-valine), and Calamendiol. These metabolites have been associated with oxidative stress, inflammation, and cardiovascular health. Functional pathway enrichment analysis revealed numerous amino acid degradation pathways, especially branched-chain amino acids, and fatty acid degradation that changed throughout the postprandial time course. CONCLUSIONS These findings suggest that CON and GRA cattle-feeding systems differentially impact whole beef metabolomics, as well as consumer postprandial metabolic responses and the associated health implications.
Collapse
Affiliation(s)
- Meghan Spears
- Department of Food Systems, Nutrition, and Kinesiology, Montana State University, Bozeman, MT 59717, USA;
| | - Gwendolyn Cooper
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA; (G.C.)
| | - Brett Sather
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA; (G.C.)
| | - Marguerite Bailey
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA; (G.C.)
| | - Jane A. Boles
- Department of Animal and Range Sciences, Montana State University, Bozeman, MT 59717, USA
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA; (G.C.)
| | - Mary P. Miles
- Department of Food Systems, Nutrition, and Kinesiology, Montana State University, Bozeman, MT 59717, USA;
| |
Collapse
|
32
|
Newton-Tanzer E, Can SN, Demmelmair H, Horak J, Holdt L, Koletzko B, Grote V. Apparent Saturation of Branched-Chain Amino Acid Catabolism After High Dietary Milk Protein Intake in Healthy Adults. J Clin Endocrinol Metab 2024:dgae599. [PMID: 39302872 DOI: 10.1210/clinem/dgae599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Indexed: 09/22/2024]
Abstract
CONTEXT Milk protein contains high concentrations of branched-chain amino acids (BCAA) that play a critical role in anabolism and are implicated in the onset of obesity and chronic disease. Characterizing BCAA catabolism in the postprandial phase could elucidate the impact of protein intake on obesity risk established in the "early protein hypothesis." OBJECTIVE To examine the acute effects of protein content of young child formulas as test meals on BCAA catabolism, observing postprandial plasma concentrations of BCAA in relation to their degradation products. METHODS The TOMI Add-On Study is a randomized, double-blind crossover study in which 27 healthy adults consumed 2 isocaloric young child formulas with alternating higher (HP) and lower (LP) protein and fat content as test meals during separate interventions, while 9 blood samples were obtained over 5 hours. BCAA, branched-chain α-keto acids (BCKA), and acylcarnitines were analyzed using a fully targeted HPLC-ESI-MS/MS approach. RESULTS Mean concentrations of BCAA, BCKA, and acylcarnitines were significantly higher after HP than LP over the 5 postprandial hours, except for the BCKA α-ketoisovalerate (KIVA). The latter metabolite showed higher postprandial concentrations after LP. With increasing mean concentrations of BCAA, concentrations of corresponding BCKA, acylcarnitines, and urea increased until a breakpoint was reached, after which concentrations of degradation products decreased (for all metabolites except valine and KIVA and Carn C4:0-iso). CONCLUSION BCAA catabolism is markedly influenced by protein content of the test meal. We present novel evidence for the apparent saturation of the BCAA degradation pathway in the acute postprandial phase up to 5 hours after consumption.
Collapse
Affiliation(s)
- Emily Newton-Tanzer
- Division of Metabolic and Nutritional Medicine, Department Paediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, and the German Center for Child and Adolescent Health, site Munich, 80337 Munich, Germany
| | - Sultan Nilay Can
- Division of Metabolic and Nutritional Medicine, Department Paediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, and the German Center for Child and Adolescent Health, site Munich, 80337 Munich, Germany
| | - Hans Demmelmair
- Division of Metabolic and Nutritional Medicine, Department Paediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, and the German Center for Child and Adolescent Health, site Munich, 80337 Munich, Germany
| | - Jeannie Horak
- Division of Metabolic and Nutritional Medicine, Department Paediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, and the German Center for Child and Adolescent Health, site Munich, 80337 Munich, Germany
| | - Lesca Holdt
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, 80337 Munich, Germany
| | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Department Paediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, and the German Center for Child and Adolescent Health, site Munich, 80337 Munich, Germany
| | - Veit Grote
- Division of Metabolic and Nutritional Medicine, Department Paediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, and the German Center for Child and Adolescent Health, site Munich, 80337 Munich, Germany
| |
Collapse
|
33
|
Romaldini A, Spanò R, Veronesi M, Grimaldi B, Bandiera T, Sabella S. Human Multi-Lineage Liver Organoid Model Reveals Impairment of CYP3A4 Expression upon Repeated Exposure to Graphene Oxide. Cells 2024; 13:1542. [PMID: 39329726 PMCID: PMC11429598 DOI: 10.3390/cells13181542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Three-dimensional hepatic cell cultures can provide an important advancement in the toxicity assessment of nanomaterials with respect to 2D models. Here, we describe liver organoids (LOs) obtained by assembling multiple cell lineages in a fixed ratio 1:1:0.2. These are upcyte® human hepatocytes, UHHs, upcyte® liver sinusoidal endothelial cells, LSECs, and human bone marrow-derived mesenchymal stromal cells, hbmMSCs. The structural and functional analyses indicated that LOs reached size stability upon ca. 10 days of cultivation (organoid maturation), showing a surface area of approximately 10 mm2 and the hepatic cellular lineages, UHHs and LSECs, arranged to form both primitive biliary networks and sinusoid structures, alike in vivo. LOs did not show signs of cellular apoptosis, senescence, or alteration of hepatocellular functions (e.g., dis-regulation of CYP3A4 or aberrant production of Albumin) for the entire culture period (19 days since organoid maturation). After that, LOs were repeatedly exposed for 19 days to a single or repeated dose of graphene oxide (GO: 2-40 µg/mL). We observed that the treatment did not induce any macroscopic signs of tissue damage, apoptosis activation, and alteration of cell viability. However, in the repeated dose regimen, we observed a down-regulation of CYP3A4 gene expression. Notably, these findings are in line with recent in vivo data, which report a similar impact on CYP3A4 when mice were repeatedly exposed to GO. Taken together, these findings warn of the potential detrimental effects of GO in real-life exposure (e.g., occupational scenario), where its progressive accumulation is likely expected. More in general, this study highlights that LOs formed by many cell lineages can enable repeated exposure regimens (suitable to mimic accumulation); thus, they can be suitably considered alternative or complementary in vitro systems to animal models.
Collapse
Affiliation(s)
- Alessio Romaldini
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| | - Raffaele Spanò
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| | - Marina Veronesi
- Structural Biophysics Facility, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy;
- D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Benedetto Grimaldi
- Molecular Medicine, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Tiziano Bandiera
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| | - Stefania Sabella
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| |
Collapse
|
34
|
Wadsworth BJ, Leiwe M, Minogue EA, Cunha PP, Engman V, Brombach C, Asvestis C, Sah-Teli SK, Marklund E, Karppinen P, Ruas JL, Rundqvist H, Lanner JT, Johnson RS. A 2-hydroxybutyrate-mediated feedback loop regulates muscular fatigue. eLife 2024; 12:RP92707. [PMID: 39226092 PMCID: PMC11371357 DOI: 10.7554/elife.92707] [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] [Indexed: 09/04/2024] Open
Abstract
Several metabolites have been shown to have independent and at times unexpected biological effects outside of their metabolic pathways. These include succinate, lactate, fumarate, and 2-hydroxyglutarate. 2-Hydroxybutyrate (2HB) is a byproduct of endogenous cysteine synthesis, produced during periods of cellular stress. 2HB rises acutely after exercise; it also rises during infection and is also chronically increased in a number of metabolic disorders. We show here that 2HB inhibits branched-chain aminotransferase enzymes, which in turn triggers a SIRT4-dependent shift in the compartmental abundance of protein ADP-ribosylation. The 2HB-induced decrease in nuclear protein ADP-ribosylation leads to a C/EBPβ-mediated transcriptional response in the branched-chain amino acid degradation pathway. This response to 2HB exposure leads to an improved oxidative capacity in vitro. We found that repeated injection with 2HB can replicate the improvement to oxidative capacity that occurs following exercise training. Together, we show that 2-HB regulates fundamental aspects of skeletal muscle metabolism.
Collapse
Affiliation(s)
- Brennan J Wadsworth
- Department of Cell and Molecular Biology, Karolinska InstituteStockholmSweden
| | - Marina Leiwe
- Department of Cell and Molecular Biology, Karolinska InstituteStockholmSweden
| | - Eleanor A Minogue
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridgeUnited Kingdom
| | - Pedro P Cunha
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridgeUnited Kingdom
| | - Viktor Engman
- Department of Physiology and Pharmacology, Karolinska InstituteStockholmSweden
| | - Carolin Brombach
- Department of Cell and Molecular Biology, Karolinska InstituteStockholmSweden
| | - Christos Asvestis
- Department of Cell and Molecular Biology, Karolinska InstituteStockholmSweden
| | - Shiv K Sah-Teli
- Faculty of Medical Biochemistry and Molecular Biology, University of OuluOuluFinland
- Department of Biochemistry, University of CambridgeCambridgeUnited Kingdom
| | - Emilia Marklund
- Department of Cell and Molecular Biology, Karolinska InstituteStockholmSweden
| | - Peppi Karppinen
- Faculty of Medical Biochemistry and Molecular Biology, University of OuluOuluFinland
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska InstituteStockholmSweden
| | - Helene Rundqvist
- Department of Laboratory Medicine, Karolinska InstitutetStockholmSweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska InstituteStockholmSweden
| | - Randall S Johnson
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
35
|
Wyatt EC, VanDerStad LR, Cook NE, McGovern MR, Zaman T, Lundin PM, Vaughan RA. Valsartan Rescues Suppressed Mitochondrial Metabolism during Insulin Resistance in C2C12 Myotubes. Cell Biochem Funct 2024; 42:e4117. [PMID: 39243192 DOI: 10.1002/cbf.4117] [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] [Revised: 08/20/2024] [Accepted: 08/25/2024] [Indexed: 09/09/2024]
Abstract
Elevated circulating branched-chain amino acids (BCAA) have been linked with the severity of insulin resistance across numerous populations, implicating heightened BCAA metabolism as a potential therapy for insulin resistance. Recently, the angiotensin II type 1 receptor (AT1R) inhibitor Valsartan (VAL) was identified as a potent inhibitor of branched-chain alpha-keto acid dehydrogenase kinase (BCKDK), a negative regulator of BCAA metabolism. This work investigated the effect of VAL on myotube metabolism and insulin sensitivity under both insulin sensitive and insulin resistant conditions. C2C12 myotubes were treated with or without VAL at 8 µM for 24 h, both with and without hyperinsulinemic-induced insulin resistance. Oxygen consumption and extracellular acidification were used to measure mitochondrial and glycolytic metabolism, respectively. Gene expression was assessed via qRT-PCR, and insulin sensitivity was assessed via Western blot. Insulin resistance significantly reduced both basal and peak mitochondrial function which were rescued to control levels by concurrent VAL. Changes in mitochondrial function occurred without substantial changes in mitochondrial content or related gene expression. Insulin sensitivity and glycolytic metabolism were unaffected by VAL, as was lipogenic signaling and lipid content. Additionally, both VAL and insulin resistance depressed Bckdha expression. Interestingly, an interaction effect was observed for extracellular isoleucine, valine, and total BCAA (but not leucine), suggesting VAL may alter BCAA utilization in an insulin sensitivity-dependent manner. Insulin resistance appears to suppress mitochondrial function in a myotube model which can be rescued by VAL. Further research will be required to explore the implications of these findings in more complex models.
Collapse
Affiliation(s)
- Emily C Wyatt
- Department of Health and Human Performance, High Point University, High Point, North Carolina, USA
| | - Lindsey R VanDerStad
- Department of Health and Human Performance, High Point University, High Point, North Carolina, USA
| | - Norah E Cook
- Department of Health and Human Performance, High Point University, High Point, North Carolina, USA
| | - Macey R McGovern
- Department of Health and Human Performance, High Point University, High Point, North Carolina, USA
| | - Toheed Zaman
- Department of Chemistry, High Point University, High Point, North Carolina, USA
| | - Pamela M Lundin
- Department of Chemistry, High Point University, High Point, North Carolina, USA
| | - Roger A Vaughan
- Department of Health and Human Performance, High Point University, High Point, North Carolina, USA
| |
Collapse
|
36
|
Reifenberg P, Zimmer A. Branched-chain amino acids: physico-chemical properties, industrial synthesis and role in signaling, metabolism and energy production. Amino Acids 2024; 56:51. [PMID: 39198298 PMCID: PMC11358235 DOI: 10.1007/s00726-024-03417-2] [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: 06/13/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Branched-chain amino acids (BCAAs)-leucine (Leu), isoleucine (Ile), and valine (Val)-are essential nutrients with significant roles in protein synthesis, metabolic regulation, and energy production. This review paper offers a detailed examination of the physico-chemical properties of BCAAs, their industrial synthesis, and their critical functions in various biological processes. The unique isomerism of BCAAs is presented, focusing on analytical challenges in their separation and quantification as well as their solubility characteristics, which are crucial for formulation and purification applications. The industrial synthesis of BCAAs, particularly using bacterial strains like Corynebacterium glutamicum, is explored, alongside methods such as genetic engineering aimed at enhancing production, detailing the enzymatic processes and specific precursors. The dietary uptake, distribution, and catabolism of BCAAs are reviewed as fundamental components of their physiological functions. Ultimately, their multifaceted impact on signaling pathways, immune function, and disease progression is discussed, providing insights into their profound influence on muscle protein synthesis and metabolic health. This comprehensive analysis serves as a resource for understanding both the basic and complex roles of BCAAs in biological systems and their industrial application.
Collapse
Affiliation(s)
- Philipp Reifenberg
- Merck Life Science KGaA, Upstream R&D, Frankfurter Strasse 250, 64293, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich‑Weiss‑Strasse 4, 64287, Darmstadt, Germany
| | - Aline Zimmer
- Merck Life Science KGaA, Upstream R&D, Frankfurter Strasse 250, 64293, Darmstadt, Germany.
| |
Collapse
|
37
|
Pi Y, Fang M, Li Y, Cai L, Han R, Sun W, Jiang X, Chen L, Du J, Zhu Z, Li X. Interactions between Gut Microbiota and Natural Bioactive Polysaccharides in Metabolic Diseases: Review. Nutrients 2024; 16:2838. [PMID: 39275156 PMCID: PMC11397228 DOI: 10.3390/nu16172838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
The gut microbiota constitutes a complex ecosystem, comprising trillions of microbes that have co-evolved with their host over hundreds of millions of years. Over the past decade, a growing body of knowledge has underscored the intricate connections among diet, gut microbiota, and human health. Bioactive polysaccharides (BPs) from natural sources like medicinal plants, seaweeds, and fungi have diverse biological functions including antioxidant, immunoregulatory, and metabolic activities. Their effects are closely tied to the gut microbiota, which metabolizes BPs into health-influencing compounds. Understanding how BPs and gut microbiota interact is critical for harnessing their potential health benefits. This review provides an overview of the human gut microbiota, focusing on its role in metabolic diseases like obesity, type II diabetes mellitus, non-alcoholic fatty liver disease, and cardiovascular diseases. It explores the basic characteristics of several BPs and their impact on gut microbiota. Given their significance for human health, we summarize the biological functions of these BPs, particularly in terms of immunoregulatory activities, blood sugar, and hypolipidemic effect, thus providing a valuable reference for understanding the potential benefits of natural BPs in treating metabolic diseases. These properties make BPs promising agents for preventing and treating metabolic diseases. The comprehensive understanding of the mechanisms by which BPs exert their effects through gut microbiota opens new avenues for developing targeted therapies to improve metabolic health.
Collapse
Affiliation(s)
- Yu Pi
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Miaoyu Fang
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Yanpin Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Long Cai
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Ruyi Han
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Wenjuan Sun
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xianren Jiang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Liang Chen
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Jun Du
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Zhigang Zhu
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Xilong Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
38
|
Mann G, Mora S, Adegoke OAJ. KIC (ketoisocaproic acid) and leucine have divergent effects on tissue insulin signaling but not on whole-body insulin sensitivity in rats. PLoS One 2024; 19:e0309324. [PMID: 39163364 PMCID: PMC11335129 DOI: 10.1371/journal.pone.0309324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/06/2024] [Indexed: 08/22/2024] Open
Abstract
Plasma levels of branched-chain amino acids and their metabolites, the branched-chain ketoacids are increased in insulin resistance. Our previous studies showed that leucine and its metabolite KIC suppress insulin-stimulated glucose uptake in L6 myotubes along with the activation of the S6K1-IRS-1 pathway. Because other tissue and fiber types can be differentially regulated by KIC, we analyzed the effect of KIC gavage on whole-body insulin sensitivity and insulin signaling in vivo. We hypothesized that KIC gavage would reduce whole-body insulin sensitivity and increase S6K1-IRS-1 phosphorylation in various tissues and muscle fibers. Five-week-old male Sprague-Dawley rats were starved for 24 hours and then gavaged with 0.75ml/100g of water, leucine (22.3g/L) or KIC (30g/L) twice, ten minutes apart. They were then euthanized at different time points post-gavage (0.5-3h), and muscle, liver, and heart tissues were dissected. Other sets of gavaged animals underwent an insulin tolerance test. Phosphorylation (ph) of S6K1 (Thr389), S6 (Ser235/6) and IRS-1 (Ser612) was increased at 30 minutes post leucine gavage in skeletal muscles irrespective of fiber type. Ph-S6 (Ser235/6) was also increased in liver and heart 30 minutes after leucine gavage. KIC gavage increased ph-S6 (Ser235/6) in the liver. Neither Leucine nor KIC influenced whole-body insulin tolerance, nor ph-Akt (Ser473) in skeletal muscle and heart. BCKD-E1 α abundance was highest in the heart and liver, while ph-BCKD-E1 α (Ser293) was higher in the gastrocnemius and EDL compared to the soleus. Our data suggests that only leucine activates the S6K1-IRS-1 signaling axis in skeletal muscle, liver and heart, while KIC only does so in the liver. The effect of leucine and KIC on the S6K1-IRS-1 signaling pathway is uncoupled from whole-body insulin sensitivity. These results suggest that KIC and leucine may not induce insulin resistance, and the contributions of other tissues may regulate whole-body insulin sensitivity in response to leucine/KIC gavage.
Collapse
Affiliation(s)
- Gagandeep Mann
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Stephen Mora
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Olasunkanmi A. John Adegoke
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| |
Collapse
|
39
|
Janovsky CCPS, Meneghini V, Tebar W, Martins JRM, Sgarbi JA, Teixeira PDFDS, Jones SR, Blaha MJ, Toth PP, Lotufo PA, Bittencourt MS, Santos RD, Santos IS, Chaker L, Bensenor IM. Branched-Chain Amino Acids, Alanine, and Thyroid Function: A Cross-Sectional, Nuclear Magnetic Resonance (NMR)-Based Approach from ELSA-Brasil. Metabolites 2024; 14:437. [PMID: 39195533 DOI: 10.3390/metabo14080437] [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: 05/15/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
The association of thyroid function with essential and non-essential amino acids is understudied, despite their common metabolic roles. Thus, our aim was to evaluate the association of thyroid function with the levels of branched-chain amino acids (BCAAs-leucine, isoleucine, and valine) and of alanine in the general population. We utilized data from the São Paulo research center of ELSA-Brasil, a longitudinal population-based cohort study. Thyroid parameters included thyroid stimulating hormone (TSH), free T4 and free T3 levels, and the FT4:FT3 ratio. BCAAs and alanine were analyzed on a fully automated NMR platform. The current analysis included euthyroid participants and participants with subclinical hyperthyroidism and hypothyroidism. We used Pearson's coefficient to quantify the correlation between thyroid-related parameters and amino acids. Linear regression models were performed to analyze whether thyroid parameters were associated with BCAAs and alanine levels. We included 4098 participants (51.3 ± 9.0 years old, 51.5% women) in this study. In the most adjusted model, higher levels of TSH were associated with higher levels of alanine, FT4 levels were inversely associated with isoleucine levels, FT3 levels were statistically significant and positively associated with valine and leucine, and the T3:T4 ratio was positively associated with all amino acids. We observed that subclinical hypothyroidism was positively associated with isoleucine and alanine levels in all models, even after full adjustment. Our findings highlight the association of subclinical hypothyroidism and thyroid-related parameters (including TSH, free T4, free T3, and FT4:FT3 ratio) with BCAAs and alanine. Further studies are needed to explore the mechanisms underlying this association. These insights contribute to our understanding of the influence of thyroid-related parameters on BCAA and alanine metabolism.
Collapse
Grants
- grants of baseline: 01 06 0010.00 RS, 01 06 0212.00 BA, 01 06 0300.00 ES, 01 06 0278.00 MG, 01 06 0115.00 SP, 01 06 0071.00 RJ; and grants of 4-year follow-up 01 10 0643-03 RS, 01 10 0742-00 BA, 01 12 0284-00 ES, 01 10 0746-00 MG, 01 10 0773-00 SP, and 01 National Council for Scientific and Technological Development
- 2015/17213- 2 Fundação de Amparo à Pesquisa do Estado de São Paulo
Collapse
Affiliation(s)
- Carolina Castro Porto Silva Janovsky
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo 05508-000, Brazil
- Division of Endocrinology, Federal University of São Paulo/Escola Paulista de Medicina, São Paulo 04039-032, Brazil
| | - Vandrize Meneghini
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo 05508-000, Brazil
| | - William Tebar
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo 05508-000, Brazil
| | - Joao Roberto Maciel Martins
- Division of Endocrinology, Federal University of São Paulo/Escola Paulista de Medicina, São Paulo 04039-032, Brazil
| | - José Augusto Sgarbi
- Division of Endocrinology, Faculty of Medicine of Marília (FAMEMA), Marília 17519-030, Brazil
| | | | - Steven R Jones
- Johns Hopkins, Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD 21287, USA
| | - Michael J Blaha
- Johns Hopkins, Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD 21287, USA
| | - Peter P Toth
- CGH Medical Center, Department of Preventive Cardiology, Sterling, IL 61081, USA
| | - Paulo A Lotufo
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo 05508-000, Brazil
| | - Marcio S Bittencourt
- Department of Medicine and Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Raul D Santos
- Heart Institute (InCor), University of São Paulo, São Paulo 05403-900, Brazil
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil
| | - Itamar S Santos
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo 05508-000, Brazil
| | - Layal Chaker
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Isabela M Bensenor
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
40
|
Kang YJ, Song W, Lee SJ, Choi SA, Chae S, Yoon BR, Kim HY, Lee JH, Kim C, Cho JY, Kim HJ, Lee WW. Inhibition of BCAT1-mediated cytosolic leucine metabolism regulates Th17 responses via the mTORC1-HIF1α pathway. Exp Mol Med 2024; 56:1776-1790. [PMID: 39085353 PMCID: PMC11372109 DOI: 10.1038/s12276-024-01286-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: 11/30/2023] [Revised: 03/26/2024] [Accepted: 04/19/2024] [Indexed: 08/02/2024] Open
Abstract
Branched-chain amino acids (BCAAs), particularly leucine, are indispensable AAs for immune regulation through metabolic rewiring. However, the molecular mechanism underlying this phenomenon remains unclear. Our investigation revealed that T-cell receptor (TCR)-activated human CD4+ T cells increase the expression of BCAT1, a cytosolic enzyme responsible for BCAA catabolism, and SLC7A5, a major BCAA transporter. This upregulation facilitates increased leucine influx and catabolism, which are particularly crucial for Th17 responses. Activated CD4+ T cells induce an alternative pathway of cytosolic leucine catabolism, generating a pivotal metabolite, β-hydroxy β-methylbutyric acid (HMB), by acting on BCAT1 and 4-hydroxyphenylpyruvate dioxygenase (HPD)/HPD-like protein (HPDL). Inhibition of BCAT1-mediated cytosolic leucine metabolism, either with BCAT1 inhibitor 2 (Bi2) or through BCAT1, HPD, or HPDL silencing using shRNA, attenuates IL-17 production, whereas HMB supplementation abrogates this effect. Mechanistically, HMB contributes to the regulation of the mTORC1-HIF1α pathway, a major signaling pathway for IL-17 production, by increasing the mRNA expression of HIF1α. This finding was corroborated by the observation that treatment with L-β-homoleucine (LβhL), a leucine analog and competitive inhibitor of BCAT1, decreased IL-17 production by TCR-activated CD4+ T cells. In an in vivo experimental autoimmune encephalomyelitis (EAE) model, blockade of BCAT1-mediated leucine catabolism, either through a BCAT1 inhibitor or LβhL treatment, mitigated EAE severity by decreasing HIF1α expression and IL-17 production in spinal cord mononuclear cells. Our findings elucidate the role of BCAT1-mediated cytoplasmic leucine catabolism in modulating IL-17 production via HMB-mediated regulation of mTORC1-HIF1α, providing insights into its relevance to inflammatory conditions.
Collapse
Affiliation(s)
- Yeon Jun Kang
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Woorim Song
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Su Jeong Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Seung Ah Choi
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Sihyun Chae
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University, College of Medicine and Hospital, Seoul, 03080, Republic of Korea
| | - Bo Ruem Yoon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Hee Young Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jung Ho Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Korea
| | - Chulwoo Kim
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Joo-Youn Cho
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University, College of Medicine and Hospital, Seoul, 03080, Republic of Korea
| | - Hyun Je Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Korea
| | - Won-Woo Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Seoul National University Cancer Research Institute, Institue of Endemic Diseases and Ischemic/Hypoxic Disease Institute, Seoul National University Medical Research Center, Seoul National University Hospital Biomedical Research Institute, Seoul, 03080, Republic of Korea.
| |
Collapse
|
41
|
Yang Q, Wang X, Liu Y, Liu J, Zhu D. Metabolic factors are not the direct mediators of the association between type 2 diabetes and osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1404747. [PMID: 39119008 PMCID: PMC11306037 DOI: 10.3389/fendo.2024.1404747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Objective The causal relationship between type 2 diabetes mellitus (T2DM) and osteoporosis (OS) remains unclear. This study aims to investigate the causal relationship and explore the potential metabolic mechanism and its mediating role. Methods We conducted a comprehensive study, gathering data on 490,089 T2DM patients from the genome-wide association study (GWAS) database and selecting OS data from FinnGen and MRC-IEU sources, including 212,778 and 463,010 patients, respectively, for causal analysis. Simultaneously, we explored the potential roles of three obesity traits and 30 metabolic and inflammation-related mediating variables in the causal relationship. Results There is a strong causal relationship between T2DM and OS. The data from our two different database sources appeared in the same direction, but after correcting for body mass index (BMI), waist circumference (WC), and waist-to-hip ratio (WHR), the direction became the same. T2DM may increase the risk of OS [odds ratio (OR) > 1.5, p < 0.001]. Steiger's test results show that there is no reverse causality. No risk factors related to glycolipid metabolism, amino acid metabolism, and inflammation were found to mediate the causal relationship. Conclusion This study's findings indicate a robust causal relationship between T2DM and OS, influenced by relevant factors such as BMI. Our results shed light on the pathogenesis of OS and underscore the importance for clinicians to treat metabolic disorders to prevent osteoporosis.
Collapse
Affiliation(s)
- Qifan Yang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Xinyu Wang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Yanwei Liu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Jing Liu
- Department of Gynecology and Obstetrics, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Dong Zhu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
42
|
Fernicola J, Vyavahare S, Gupta SK, Kalwaghe A, Kosmac K, Davis A, Nicholson M, Isales CM, Shinde R, Fulzele S. The Role of Branched Chain Ketoacid Dehydrogenase Kinase (BCKDK) in Skeletal Muscle Biology and Pathogenesis. Int J Mol Sci 2024; 25:7601. [PMID: 39062842 PMCID: PMC11277350 DOI: 10.3390/ijms25147601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Muscle wasting can be caused by nutrition deficiency and inefficient metabolism of amino acids, including Branched Chain Amino Acids (BCAAs). Branched Chain Amino Acids are a major contributor to the metabolic needs of healthy muscle and account for over a tenth of lean muscle mass. Branched chain alpha-ketoacid dehydrogenase (BCKD) is the rate limiting enzyme of BCAA metabolism. Inhibition of BCKD is achieved through a reversible phosphorylation event by Branched Chain a-ketoacid dehydrogenase kinase (BCKDK). Our study set out to determine the importance of BCKDK in the maintenance of skeletal muscle. We used the Gene Expression Omnibus Database to understand the role of BCKDK in skeletal muscle pathogenesis, including aging, muscular disease, and interrupted muscle metabolism. We found BCKDK expression levels were consistently decreased in pathologic conditions. These results were most consistent when exploring muscular disease followed by aging. Based on our findings, we hypothesize that decreased BCKDK expression alters BCAA catabolism and impacts loss of normal muscle integrity and function. Further research could offer valuable insights into potential therapeutic strategies for addressing muscle-related disorders.
Collapse
Affiliation(s)
- Joshua Fernicola
- Department of Medicine, Division of Endocrinology, Augusta University, Augusta, GA 30912, USA
| | - Sagar Vyavahare
- Department of Medicine, Division of Endocrinology, Augusta University, Augusta, GA 30912, USA
| | - Sonu Kumar Gupta
- Department of Medicine, Division of Endocrinology, Augusta University, Augusta, GA 30912, USA
| | - Aditya Kalwaghe
- Department of Medicine, Division of Endocrinology, Augusta University, Augusta, GA 30912, USA
| | - Kate Kosmac
- Department of Physical Therapy, Augusta University, Augusta, GA 30912, USA
| | - Adam Davis
- Department of Medicine, Division of Endocrinology, Augusta University, Augusta, GA 30912, USA
| | - Matthew Nicholson
- Department of Medicine, Division of Endocrinology, Augusta University, Augusta, GA 30912, USA
| | - Carlos M Isales
- Department of Medicine, Division of Endocrinology, Augusta University, Augusta, GA 30912, USA
- Center for Healthy Aging, Augusta University, Augusta, GA 30912, USA
| | - Rahul Shinde
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute Cancer Center, Philadelphia, PA 19104, USA
| | - Sadanand Fulzele
- Department of Medicine, Division of Endocrinology, Augusta University, Augusta, GA 30912, USA
- Center for Healthy Aging, Augusta University, Augusta, GA 30912, USA
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
43
|
Parvanovova P, Hnilicova P, Kolisek M, Tatarkova Z, Halasova E, Kurca E, Holubcikova S, Koprusakova MT, Baranovicova E. Disturbances in Muscle Energy Metabolism in Patients with Amyotrophic Lateral Sclerosis. Metabolites 2024; 14:356. [PMID: 39057679 PMCID: PMC11278632 DOI: 10.3390/metabo14070356] [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: 05/22/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disease type of motor neuron disorder characterized by degeneration of the upper and lower motor neurons resulting in dysfunction of the somatic muscles of the body. The ALS condition is manifested in progressive skeletal muscle atrophy and spasticity. It leads to death, mostly due to respiratory failure. Within the pathophysiology of the disease, muscle energy metabolism seems to be an important part. In our study, we used blood plasma from 25 patients with ALS diagnosed by definitive El Escorial criteria according to ALSFR-R (Revised Amyotrophic Lateral Sclerosis Functional Rating Scale) criteria and 25 age and sex-matched subjects. Aside from standard clinical biochemical parameters, we used the NMR (nuclear magnetic resonance) metabolomics approach to determine relative plasma levels of metabolites. We observed a decrease in total protein level in blood; however, despite accelerated skeletal muscle catabolism characteristic for ALS patients, we did not detect changes in plasma levels of essential amino acids. When focused on alterations in energy metabolism within muscle, compromised creatine uptake was accompanied by decreased plasma creatinine. We did not observe changes in plasma levels of BCAAs (branched chain amino acids; leucine, isoleucine, valine); however, the observed decrease in plasma levels of all three BCKAs (branched chain alpha-keto acids derived from BCAAs) suggests enhanced utilization of BCKAs as energy substrate. Glutamine, found to be increased in blood plasma in ALS patients, besides serving for ammonia detoxification, could also be considered a potential TCA (tricarboxylic acid) cycle contributor in times of decreased pyruvate utilization. When analyzing the data by using a cross-validated Random Forest algorithm, it finished with an AUC of 0.92, oob error of 8%, and an MCC (Matthew's correlation coefficient) of 0.84 when relative plasma levels of metabolites were used as input variables. Although the discriminatory power of the system used was promising, additional features are needed to create a robust discriminatory model.
Collapse
Affiliation(s)
- Petra Parvanovova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.P.); (Z.T.); (S.H.)
| | - Petra Hnilicova
- Biomedical Centre Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.H.); (M.K.); (E.H.)
| | - Martin Kolisek
- Biomedical Centre Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.H.); (M.K.); (E.H.)
| | - Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.P.); (Z.T.); (S.H.)
| | - Erika Halasova
- Biomedical Centre Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.H.); (M.K.); (E.H.)
| | - Egon Kurca
- Department of Neurology, University Hospital Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia;
| | - Simona Holubcikova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.P.); (Z.T.); (S.H.)
| | - Monika Turcanova Koprusakova
- Department of Neurology, University Hospital Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia;
| | - Eva Baranovicova
- Biomedical Centre Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.H.); (M.K.); (E.H.)
| |
Collapse
|
44
|
Cook NE, McGovern MR, Zaman T, Lundin PM, Vaughan RA. Effect of mTORC Agonism via MHY1485 with and without Rapamycin on C2C12 Myotube Metabolism. Int J Mol Sci 2024; 25:6819. [PMID: 38999929 PMCID: PMC11241331 DOI: 10.3390/ijms25136819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024] Open
Abstract
The mechanistic target of rapamycin complex (mTORC) regulates protein synthesis and can be activated by branched-chain amino acids (BCAAs). mTORC has also been implicated in the regulation of mitochondrial metabolism and BCAA catabolism. Some speculate that mTORC overactivation by BCAAs may contribute to insulin resistance. The present experiments assessed the effect of mTORC activation on myotube metabolism and insulin sensitivity using the mTORC agonist MHY1485, which does not share structural similarities with BCAAs. METHODS C2C12 myotubes were treated with MHY1485 or DMSO control both with and without rapamycin. Gene expression was assessed using qRT-PCR and insulin sensitivity and protein expression by western blot. Glycolytic and mitochondrial metabolism were measured by extracellular acidification rate and oxygen consumption. Mitochondrial and lipid content were analyzed by fluorescent staining. Liquid chromatography-mass spectrometry was used to assess extracellular BCAAs. RESULTS Rapamycin reduced p-mTORC expression, mitochondrial content, and mitochondrial function. Surprisingly, MHY1485 did not alter p-mTORC expression or cell metabolism. Neither treatment altered indicators of BCAA metabolism or extracellular BCAA content. CONCLUSION Collectively, inhibition of mTORC via rapamycin reduces myotube metabolism and mitochondrial content but not BCAA metabolism. The lack of p-mTORC activation by MHY1485 is a limitation of these experiments and warrants additional investigation.
Collapse
Affiliation(s)
- Norah E. Cook
- Department of Health and Human Performance, High Point University, High Point, NC 27262-3598, USA; (N.E.C.); (M.R.M.)
| | - Macey R. McGovern
- Department of Health and Human Performance, High Point University, High Point, NC 27262-3598, USA; (N.E.C.); (M.R.M.)
| | - Toheed Zaman
- Department of Chemistry, High Point University, High Point, NC 27262-3598, USA; (T.Z.); (P.M.L.)
| | - Pamela M. Lundin
- Department of Chemistry, High Point University, High Point, NC 27262-3598, USA; (T.Z.); (P.M.L.)
| | - Roger A. Vaughan
- Department of Health and Human Performance, High Point University, High Point, NC 27262-3598, USA; (N.E.C.); (M.R.M.)
| |
Collapse
|
45
|
Tanase DM, Valasciuc E, Costea CF, Scripcariu DV, Ouatu A, Hurjui LL, Tarniceriu CC, Floria DE, Ciocoiu M, Baroi LG, Floria M. Duality of Branched-Chain Amino Acids in Chronic Cardiovascular Disease: Potential Biomarkers versus Active Pathophysiological Promoters. Nutrients 2024; 16:1972. [PMID: 38931325 PMCID: PMC11206939 DOI: 10.3390/nu16121972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Branched-chain amino acids (BCAAs), comprising leucine (Leu), isoleucine (Ile), and valine (Val), are essential nutrients vital for protein synthesis and metabolic regulation via specialized signaling networks. Their association with cardiovascular diseases (CVDs) has become a focal point of scientific debate, with emerging evidence suggesting both beneficial and detrimental roles. This review aims to dissect the multifaceted relationship between BCAAs and cardiovascular health, exploring the molecular mechanisms and clinical implications. Elevated BCAA levels have also been linked to insulin resistance (IR), type 2 diabetes mellitus (T2DM), inflammation, and dyslipidemia, which are well-established risk factors for CVD. Central to these processes are key pathways such as mammalian target of rapamycin (mTOR) signaling, nuclear factor kappa-light-chain-enhancer of activate B cells (NF-κB)-mediated inflammation, and oxidative stress. Additionally, the interplay between BCAA metabolism and gut microbiota, particularly the production of metabolites like trimethylamine-N-oxide (TMAO), adds another layer of complexity. Contrarily, some studies propose that BCAAs may have cardioprotective effects under certain conditions, contributing to muscle maintenance and metabolic health. This review critically evaluates the evidence, addressing the biological basis and signal transduction mechanism, and also discusses the potential for BCAAs to act as biomarkers versus active mediators of cardiovascular pathology. By presenting a balanced analysis, this review seeks to clarify the contentious roles of BCAAs in CVD, providing a foundation for future research and therapeutic strategies required because of the rising prevalence, incidence, and total burden of CVDs.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Claudia Florida Costea
- Department of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- 2nd Ophthalmology Clinic, “Prof. Dr. Nicolae Oblu” Emergency Clinical Hospital, 700309 Iași, Romania
| | - Dragos Viorel Scripcariu
- Department of General Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Regional Institute of Oncology, 700483 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Loredana Liliana Hurjui
- Department of Morpho-Functional Sciences II, Physiology Discipline, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Laboratory, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Claudia Cristina Tarniceriu
- Department of Morpho-Functional Sciences I, Discipline of Anatomy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Diana Elena Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Livia Genoveva Baroi
- Department of Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Vascular Surgery, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| |
Collapse
|
46
|
Cook NE, McGovern MR, Zaman T, Lundin PM, Vaughan RA. Fructose Reduces Mitochondrial Metabolism and Increases Extracellular BCAA during Insulin Resistance in C2C12 Myotubes. Nutrients 2024; 16:1582. [PMID: 38892515 PMCID: PMC11174010 DOI: 10.3390/nu16111582] [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: 04/22/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
Fructose is a commonly consumed monosaccharide implicated in developing several metabolic diseases. Previously, elevated branched-chain amino acids (BCAA) have been correlated with the severity of insulin resistance. Most recently, the effect of fructose consumption on the downregulation of BCAA catabolic enzymes was observed. Thus, this mechanistic study investigated the effects of physiologically attainable levels of fructose, both with and without concurrent insulin resistance, in a myotube model of skeletal muscle. METHODS C2C12 mouse myoblasts were treated with fructose at a concentration of 100 µM (which approximates physiologically attainable concentrations in peripheral circulation) both with and without hyperinsulinemic-mediated insulin resistance. Gene expression was assessed by qRT-PCR, and protein expression was assessed by Western blot. Oxygen consumption rate and extracellular acidification rate were used to assess mitochondrial oxidative and glycolytic metabolism, respectively. Liquid chromatography-mass spectrometry was utilized to analyze leucine, isoleucine and valine concentration values. RESULTS Fructose significantly reduced peak glycolytic and peak mitochondrial metabolism without altering related gene or protein expression. Similarly, no effect of fructose on BCAA catabolic enzymes was observed; however, fructose treatment resulted in elevated total extracellular BCAA in insulin-resistant cells. DISCUSSION Collectively, these observations demonstrate that fructose at physiologically attainable levels does not appear to alter insulin sensitivity or BCAA catabolic potential in cultured myotubes. However, fructose may depress peak cell metabolism and BCAA utilization during insulin resistance.
Collapse
Affiliation(s)
- Norah E. Cook
- Department of Health and Human Performance, High Point University, One University Parkway, High Point, NC 27268, USA; (N.E.C.); (M.R.M.)
| | - Macey R. McGovern
- Department of Health and Human Performance, High Point University, One University Parkway, High Point, NC 27268, USA; (N.E.C.); (M.R.M.)
| | - Toheed Zaman
- Department of Chemistry, High Point University, High Point, NC 27268, USA; (T.Z.); (P.M.L.)
| | - Pamela M. Lundin
- Department of Chemistry, High Point University, High Point, NC 27268, USA; (T.Z.); (P.M.L.)
| | - Roger A. Vaughan
- Department of Health and Human Performance, High Point University, One University Parkway, High Point, NC 27268, USA; (N.E.C.); (M.R.M.)
| |
Collapse
|
47
|
Jang JH, Joung JY, Pack SP, Oh NS. Preventive effect of fermented whey protein mediated by Lactobacillus gasseri IM13 via the PI3K/AKT/FOXO pathway in muscle atrophy. J Dairy Sci 2024; 107:2606-2619. [PMID: 37977441 DOI: 10.3168/jds.2023-24027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
This study investigated the preventive effects of whey protein fermented with Lactobacillus gasseri IM13 (F-WP) against dexamethasone (DEX)-induced muscle atrophy. C2C12 muscle cells were treated with F-WP followed by DEX treatment. Dexamethasone treatment inhibited myotube formation and the expression of myogenic regulatory factors; however, pretreatment with F-WP attenuated DEX-induced damage. The F-WP significantly activated the phosphorylation of the IGF-1/PI3K/AKT pathway and improved muscle homeostasis suppressed by DEX. Moreover, F-WP alleviated the phosphorylation of mTOR, S6K1, and 4E-BP1 and enhanced muscle protein synthesis. Muscle-specific ubiquitin ligases and autophagy lysosomes, which were activated by the dephosphorylation of FOXO3a by DEX treatment, were significantly attenuated by F-WP pretreatment of myotubes. For peptidomic analysis, F-WP was fractionated using preparative HPLC (prep-HPLC), and the AA sequences of 11 peptides were identified using MALDI-TOF/MS/MS. In conclusion, fermentation of whey protein by the specific probiotic strain IM13 produced bioactive peptides with high antioxidant and anti-sarcopenic-sarcopenic effects, which markedly enhanced myogenesis and muscle protein synthesis while diminishing muscle protein degradation compared with intact whey protein.
Collapse
Affiliation(s)
- Ji Hun Jang
- Department of Food and Biotechnology, Korea University, Sejong 30019, Korea
| | - Jae Yeon Joung
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Korea
| | - Nam Su Oh
- Department of Food and Biotechnology, Korea University, Sejong 30019, Korea.
| |
Collapse
|
48
|
Fine KS, Wilkins JT, Sawicki KT. Circulating Branched Chain Amino Acids and Cardiometabolic Disease. J Am Heart Assoc 2024; 13:e031617. [PMID: 38497460 PMCID: PMC11179788 DOI: 10.1161/jaha.123.031617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Branched chain amino acids (BCAAs) are essential for protein homeostasis, energy balance, and signaling pathways. Changes in BCAA homeostasis have emerged as pivotal contributors in the pathophysiology of several cardiometabolic diseases, including type 2 diabetes, obesity, hypertension, atherosclerotic cardiovascular disease, and heart failure. In this review, we provide a detailed overview of BCAA metabolism, focus on molecular mechanisms linking disrupted BCAA homeostasis with cardiometabolic disease, summarize the evidence from observational and interventional studies investigating associations between circulating BCAAs and cardiometabolic disease, and offer valuable insights into the potential for BCAA manipulation as a novel therapeutic strategy for cardiometabolic disease.
Collapse
Affiliation(s)
- Keenan S. Fine
- Northwestern University Feinberg School of MedicineChicagoILUSA
| | - John T. Wilkins
- Northwestern University Feinberg School of MedicineChicagoILUSA
- Division of Cardiology, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoILUSA
| | - Konrad T. Sawicki
- Northwestern University Feinberg School of MedicineChicagoILUSA
- Division of Cardiology, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoILUSA
| |
Collapse
|
49
|
Mora S, Mann G, Adegoke OAJ. Sex differences in cachexia and branched-chain amino acid metabolism following chemotherapy in mice. Physiol Rep 2024; 12:e16003. [PMID: 38631892 PMCID: PMC11023815 DOI: 10.14814/phy2.16003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Chemotherapy is a major contributor to cachexia, but studies often investigate male animals. Here, we investigated whether sex modifies the effects of chemotherapy on cachexia and BCAA metabolism. Ten-week-old CD2F1 male and female mice were treated with the chemotherapy drug cocktail folfiri (50 mg/kg 5-fluorouracil, 90 mg/kg leucovorin, and 24 mg/kg CPT11) (drug) or vehicle twice a week for 6 weeks. Insulin tolerance tests were conducted and BCAA levels and metabolism were measured in plasma and tissues. Drug treatment reduced body and skeletal muscle weights and anabolic signaling in both sexes, with females showing worsened outcomes (p < 0.05 for all). Drug treatment increased plasma BCAA only in males, but BCAA concentrations in the skeletal muscle of both sexes were decreased; this decrease was more profound in males (p = 0.0097). In addition, muscle expression of the BCAA transporter LAT1 was reduced; this reduction was more severe in females (p = 0.0264). In both sexes, the (inhibitory) phosphorylation of BCKD-E1αser293 was increased along with decreased BCKD activity. In the liver, drug treatment increased BCAA concentrations and LAT1 expression, but BCKD activity was suppressed in both sexes (p < 0.05 for all). Our results demonstrate that altered BCAA metabolism may contribute to chemotherapy-induced cachexia in a sex-dependent manner.
Collapse
Affiliation(s)
- Stephen Mora
- Muscle Health Research Centre, School of Kinesiology and Health ScienceYork UniversityTorontoOntarioCanada
| | - Gagandeep Mann
- Muscle Health Research Centre, School of Kinesiology and Health ScienceYork UniversityTorontoOntarioCanada
| | - Olasunkanmi A. J. Adegoke
- Muscle Health Research Centre, School of Kinesiology and Health ScienceYork UniversityTorontoOntarioCanada
| |
Collapse
|
50
|
Lesnik C, Kaletsky R, Ashraf JM, Sohrabi S, Cota V, Sengupta T, Keyes W, Luo S, Murphy CT. Enhanced branched-chain amino acid metabolism improves age-related reproduction in C. elegans. Nat Metab 2024; 6:724-740. [PMID: 38418585 DOI: 10.1038/s42255-024-00996-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
Reproductive ageing is one of the earliest human ageing phenotypes, and mitochondrial dysfunction has been linked to oocyte quality decline; however, it is not known which mitochondrial metabolic processes are critical for oocyte quality maintenance with age. To understand how mitochondrial processes contribute to Caenorhabditis elegans oocyte quality, we characterized the mitochondrial proteomes of young and aged wild-type and long-reproductive daf-2 mutants. Here we show that the mitochondrial proteomic profiles of young wild-type and daf-2 worms are similar and share upregulation of branched-chain amino acid (BCAA) metabolism pathway enzymes. Reduction of the BCAA catabolism enzyme BCAT-1 shortens reproduction, elevates mitochondrial reactive oxygen species levels, and shifts mitochondrial localization. Moreover, bcat-1 knockdown decreases oocyte quality in daf-2 worms and reduces reproductive capability, indicating the role of this pathway in the maintenance of oocyte quality with age. Notably, oocyte quality deterioration can be delayed, and reproduction can be extended in wild-type animals both by bcat-1 overexpression and by supplementing with vitamin B1, a cofactor needed for BCAA metabolism.
Collapse
Affiliation(s)
- Chen Lesnik
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
- Faculty of Natural Sciences, Department of Human Biology, University of Haifa, Haifa, Israel
| | - Rachel Kaletsky
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Jasmine M Ashraf
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Salman Sohrabi
- LSI Genomics, Princeton University, Princeton, NJ, USA
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Vanessa Cota
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
- Department of Biology, Tacoma Community College, Tacoma, WA, USA
| | - Titas Sengupta
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - William Keyes
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Shijing Luo
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Coleen T Murphy
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- LSI Genomics, Princeton University, Princeton, NJ, USA.
| |
Collapse
|