1
|
Nguyen QD, Nguyen TVL, Tran TTV, Ngo-Thi NH, Truong-Thi HT, Nguyen VL, Luong TK, Do AD. Fermentation of Ngoc Linh ginseng (Panax vietnamensis Ha et Grushv.) root extract using Levilactobacillus brevis QD-1: Changes in majonoside R2 content and pharmacological activities. Food Res Int 2025; 208:116275. [PMID: 40263859 DOI: 10.1016/j.foodres.2025.116275] [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: 01/10/2025] [Revised: 02/19/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025]
Abstract
This study investigated the biotransform beneficial effects of Levilactobacillus brevis QD-1 fermentation on the bioactive compounds and pharmacological activities of Ngoc Linh ginseng root extract through in silico and in vitro analyses. Whole-genome sequencing revealed that L. brevis QD-1 lacks virulence factors and biogenic amine synthesis genes, confirming its safety for food and pharmaceutical applications. Genome annotation identified β-glucosidase from glycoside hydrolase families (GH1, GH2, GH3, and GH30), suggesting its potential role in ginsenoside biotransformation. In vitro study demonstrated that L. brevis QD-1 might proliferate in Ngoc Linh ginseng extract and express β-glucosidase after 24 h of fermentation. Fermentation significantly altered the ginsenoside profile, notably increasing the rare saponin majonoside R2 from 479.65 mg/L to 649.32 mg/L after 48 h. Additionally, fermentation enhanced total phenolic content and facilitated the release of essential and non-essential amino acids, particularly threonine, valine, lysine, and histidine, which were initially undetectable in the raw extract. These compositional changes correlated with improved pharmacological properties, as the fermented extract exhibited significantly stronger antioxidant, anti-inflammatory, and DNA-protective activities compared to the raw extract. These findings highlight the potential of L. brevis QD-1 as a safe and effective microbial agent for enhancing the bioactivity of Ngoc Linh ginseng, paving the way for its applications in functional foods and natural therapeutics.
Collapse
Affiliation(s)
- Quoc-Duy Nguyen
- Department of Food Technology, Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Viet Nam
| | - Thi-Van-Linh Nguyen
- Department of Food Technology, Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Viet Nam
| | - Thi Tuong Vi Tran
- Department of Food Technology, Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Viet Nam
| | | | | | - Vinh-Lam Nguyen
- Department of Food Technology, Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Viet Nam
| | | | - Anh Duy Do
- Department of Biotechnology, NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Viet Nam.
| |
Collapse
|
2
|
Devaraju M, Prasad Dasappa J. Oxidative annulation of L-phenylalanine using I 2/DMSO: an easy approach for chemoselective synthesis of 2,3,5-trisubstituted pyridines and 2,5-disubstituted oxazoles. Org Biomol Chem 2025; 23:3437-3442. [PMID: 40084585 DOI: 10.1039/d5ob00039d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
A facile approach for chemoselective synthesis of 2,3,5-trisubstituted pyridines and 2,5-disubstituted oxazoles from L-phenylalanine and aryl methyl ketone using I2/DMSO has been developed. Two equivalents of L-phenylalanine reacted with aryl methyl ketone and I2/DMSO giving 2,3,5-trisubstituted pyridines and interestingly one equivalent of L-phenylalanine afforded 2,5-disubstituted oxazoles. This chemo-divergent approach has features of being transition metal-free, free from expensive ligands, additives and starting materials, and having a less toxic nitrogen source and broad substrate scope.
Collapse
Affiliation(s)
- Mohankumar Devaraju
- Department of Chemistry, Mangalore University, Mangalagangotri-574199, Karnataka State, India.
| | | |
Collapse
|
3
|
Chen Y, Liang R, Li Y, Jiang L, Ma D, Luo Q, Song G. Chromatin accessibility: biological functions, molecular mechanisms and therapeutic application. Signal Transduct Target Ther 2024; 9:340. [PMID: 39627201 PMCID: PMC11615378 DOI: 10.1038/s41392-024-02030-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/04/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024] Open
Abstract
The dynamic regulation of chromatin accessibility is one of the prominent characteristics of eukaryotic genome. The inaccessible regions are mainly located in heterochromatin, which is multilevel compressed and access restricted. The remaining accessible loci are generally located in the euchromatin, which have less nucleosome occupancy and higher regulatory activity. The opening of chromatin is the most important prerequisite for DNA transcription, replication, and damage repair, which is regulated by genetic, epigenetic, environmental, and other factors, playing a vital role in multiple biological progresses. Currently, based on the susceptibility difference of occupied or free DNA to enzymatic cleavage, solubility, methylation, and transposition, there are many methods to detect chromatin accessibility both in bulk and single-cell level. Through combining with high-throughput sequencing, the genome-wide chromatin accessibility landscape of many tissues and cells types also have been constructed. The chromatin accessibility feature is distinct in different tissues and biological states. Research on the regulation network of chromatin accessibility is crucial for uncovering the secret of various biological processes. In this review, we comprehensively introduced the major functions and mechanisms of chromatin accessibility variation in different physiological and pathological processes, meanwhile, the targeted therapies based on chromatin dynamics regulation are also summarized.
Collapse
Affiliation(s)
- Yang Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Rui Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Yong Li
- Hepatobiliary Pancreatic Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Lingli Jiang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Di Ma
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China.
| |
Collapse
|
4
|
Goldberg D, Buchshtab N, Charni-Natan M, Goldstein I. Transcriptional cascades during fasting amplify gluconeogenesis and instigate a secondary wave of ketogenic gene transcription. Liver Int 2024; 44:2964-2982. [PMID: 39162082 DOI: 10.1111/liv.16077] [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: 03/17/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND AND AIMS During fasting, bodily homeostasis is maintained due to hepatic production of glucose (gluconeogenesis) and ketone bodies (ketogenesis). The main hormones governing hepatic fuel production are glucagon and glucocorticoids that initiate transcriptional programs aimed at supporting gluconeogenesis and ketogenesis. METHODS Using primary mouse hepatocytes as an ex vivo model, we employed transcriptomic analysis (RNA-seq), genome-wide profiling of enhancer dynamics (ChIP-seq), perturbation experiments (inhibitors, shRNA), hepatic glucose production measurements and computational analyses. RESULTS We found that in addition to the known metabolic genes transcriptionally induced by glucagon and glucocorticoids, these hormones induce a set of genes encoding transcription factors (TFs) thereby initiating transcriptional cascades. Upon activation by glucocorticoids, the glucocorticoid receptor (GR) induced the genes encoding two TFs: CCAAT/enhancer-binding protein beta (C/EBPβ) and peroxisome proliferator-activated receptor alpha (PPARα). We found that the GR-C/EBPβ cascade mainly serves as a secondary amplifier of primary hormone-induced gene programs. C/EBPβ augmented gluconeogenic gene expression and hepatic glucose production. Conversely, the GR-PPARα cascade initiated a secondary transcriptional wave of genes supporting ketogenesis. The cascade led to synergistic induction of ketogenic genes which is dependent on protein synthesis. Genome-wide analysis of enhancer dynamics revealed numerous enhancers activated by the GR-PPARα cascade. These enhancers were proximal to ketogenic genes, enriched for the PPARα response element and showed increased PPARα binding. CONCLUSION This study reveals abundant transcriptional cascades occurring during fasting. These cascades serve two separated purposes: the amplification of the gluconeogenic transcriptional program and the induction of a gene program aimed at enhancing ketogenesis.
Collapse
Affiliation(s)
- Dana Goldberg
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Nufar Buchshtab
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Meital Charni-Natan
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Ido Goldstein
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
5
|
FINK JULIUS, TANAKA MASAMI, HORIE SHIGEO. Effects of Fasting on Metabolic Hormones and Functions: A Narrative Review. JUNTENDO IJI ZASSHI = JUNTENDO MEDICAL JOURNAL 2024; 70:348-359. [PMID: 39545228 PMCID: PMC11560338 DOI: 10.14789/jmj.jmj24-0012-r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/17/2024] [Indexed: 11/17/2024]
Abstract
The occurrence of the metabolic syndrome and its related diseases such as diabetes are steadily rising in our modern society. Modern food choices and the more sedentary lifestyles largely contribute to this shift in our society's health. Fasting has been practiced for religious purposes all over the world long time before science showed the benefits of it. The effects of fasting on glucose and fat metabolism are of great interest. Fasting triggers a cascade of changes in the hormonal, microbiome and enzymatic environments, leading to shifted glucose and fat metabolisms. Fasting-induced metabolic function changes are affected by several factors such as sex hormones, lipid-released hormones, growth hormone, insulin, and the gut microbiome, leading to lipolysis and the release of FFA into the bloodstream. The purpose of this review is to summarize the newest research results on the specific pathways fasting triggers to improve metabolic functions and understand the potential applications of fasting as prevention/treatment of several metabolic conditions.
Collapse
Affiliation(s)
- JULIUS FINK
- Corresponding author: Julius Fink, Department of Urology, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan, TEL: +81-3-3813-3111 E-mail:
| | | | | |
Collapse
|
6
|
Maruszczak K, Koren P, Radzikowski K, Pixner T, Suppli MP, Wewer Albrechtsen NJ, Weghuber D, Torbahn G. Glucagon, Metabolic Dysfunction-Associated Steatotic Liver Disease and Amino Acids in Humans and Animals without Diabetes Mellitus-An Evidence Map. Life (Basel) 2024; 14:1292. [PMID: 39459592 PMCID: PMC11509797 DOI: 10.3390/life14101292] [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: 08/22/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
INTRODUCTION Health systems are confronted with not only the growing worldwide childhood obesity epidemic but also associated comorbidities. These subsequently cause variations in distinct metabolic pathways, leading to metabolic dysfunction-associated steatotic liver disease (MASLD). The aim of this evidence map is to systematically evaluate the evidence and to identify research gaps on glucagon-induced amino acid (AA) turnover and its metabolic interaction with MASLD. METHODOLOGY A systematic literature search was conducted up to April 2023 in three electronic databases. Studies were required to include at least two of the main research areas, glucagon, AA metabolism and MASLD. Two independent reviewers screened titles and abstracts according to prespecified eligibility criteria, as well as full-text articles. Results are summarized in tables stratified by human and animal studies and study population age. RESULTS Thirty-four references were ultimately included. The publication years dated back to 1965 showed a great increase from 2012 to 2023. In total, there were 19 animal studies and 15 human studies. Among the human studies, except for two studies in adolescents, all the studies were conducted in adults. In human studies, the methods used to evaluate metabolic changes differed among hyperinsulinemic-euglycemic clamp and oral glucose tolerance tests. Thirteen studies focused on the metabolic effects of MASLD, while only two studies explored the interaction between MASLD, glucagon and AA metabolism in humans. The other 19 studies focused on metabolomics, beta cell function or just one topic of a research area and not on interactions between one another. CONCLUSION Research on the interaction between MASLD, glucagon and AA metabolism in humans is sparse and complete lacking in pediatrics. Furthermore, longitudinal studies with a focus on hyperglucagonemia independent of diabetes but related to MASLD present an unambiguous research gap.
Collapse
Affiliation(s)
- Katharina Maruszczak
- Department of Pediatrics, University Hospital Salzburg, Division of Gastroenterology, Hepatology and Nutrition, Paracelsus Medical University, 5020 Salzburg, Austria; (K.M.); (P.K.); (K.R.); (G.T.)
- Obesity Research Unit, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Pia Koren
- Department of Pediatrics, University Hospital Salzburg, Division of Gastroenterology, Hepatology and Nutrition, Paracelsus Medical University, 5020 Salzburg, Austria; (K.M.); (P.K.); (K.R.); (G.T.)
| | - Konrad Radzikowski
- Department of Pediatrics, University Hospital Salzburg, Division of Gastroenterology, Hepatology and Nutrition, Paracelsus Medical University, 5020 Salzburg, Austria; (K.M.); (P.K.); (K.R.); (G.T.)
| | - Thomas Pixner
- Obesity Research Unit, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria;
- Department of Pediatric and Adolescent Medicine, Salzkammergutklinikum Voecklabruck, 4864 Voecklabruck, Austria
| | - Malte Palm Suppli
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg, 2400 Copenhagen, Denmark;
| | | | - Daniel Weghuber
- Department of Pediatrics, University Hospital Salzburg, Division of Gastroenterology, Hepatology and Nutrition, Paracelsus Medical University, 5020 Salzburg, Austria; (K.M.); (P.K.); (K.R.); (G.T.)
- Obesity Research Unit, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Gabriel Torbahn
- Department of Pediatrics, University Hospital Salzburg, Division of Gastroenterology, Hepatology and Nutrition, Paracelsus Medical University, 5020 Salzburg, Austria; (K.M.); (P.K.); (K.R.); (G.T.)
- Obesity Research Unit, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria;
- Department of Pediatrics, Paracelsus Medical University, Klinikum Nürnberg, Universitätsklinik der Paracelsus Medizinischen Privatuniversität Nürnberg, 90471 Nuremberg, Germany
| |
Collapse
|
7
|
Li H, Liu P, Zhang B, Yuan Z, Guo M, Zou X, Qian Y, Deng S, Zhu L, Cao X, Tao T, Xia S, Bao X, Xu Y. Acute ischemia induces spatially and transcriptionally distinct microglial subclusters. Genome Med 2023; 15:109. [PMID: 38082331 PMCID: PMC10712107 DOI: 10.1186/s13073-023-01257-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Damage in the ischemic core and penumbra after stroke affects patient prognosis. Microglia immediately respond to ischemic insult and initiate immune inflammation, playing an important role in the cellular injury after stroke. However, the microglial heterogeneity and the mechanisms involved remain unclear. METHODS We first performed single-cell RNA-sequencing (scRNA-seq) and spatial transcriptomics (ST) on middle cerebral artery occlusion (MCAO) mice from three time points to determine stroke-associated microglial subclusters and their spatial distributions. Furthermore, the expression of microglial subcluster-specific marker genes and the localization of different microglial subclusters were verified on MCAO mice through RNAscope and immunofluorescence. Gene set variation analysis (GSVA) was performed to reveal functional characteristics of microglia sub-clusters. Additionally, ingenuity pathway analysis (IPA) was used to explore upstream regulators of microglial subclusters, which was confirmed by immunofluorescence, RT-qPCR, shRNA-mediated knockdown, and targeted metabolomics. Finally, the infarct size, neurological deficits, and neuronal apoptosis were evaluated in MCAO mice after manipulation of specific microglial subcluster. RESULTS We discovered stroke-associated microglial subclusters in the brains of MCAO mice. We also identified novel marker genes of these microglial subclusters and defined these cells as ischemic core-associated (ICAM) and ischemic penumbra-associated (IPAM) microglia, according to their spatial distribution. ICAM, induced by damage-associated molecular patterns, are probably fueled by glycolysis, and exhibit increased pro-inflammatory cytokines and chemokines production. BACH1 is a key transcription factor driving ICAM generation. In contrast, glucocorticoids, which are enriched in the penumbra, likely trigger IPAM formation, which are presumably powered by the citrate cycle and oxidative phosphorylation and are characterized by moderate pro-inflammatory responses, inflammation-alleviating metabolic features, and myelinotrophic properties. CONCLUSIONS ICAM could induce excessive neuroinflammation, aggravating brain injury, whereas IPAM probably exhibit neuroprotective features, which could be essential for the homeostasis and survival of cells in the penumbra. Our findings provide a biological basis for targeting specific microglial subclusters as a potential therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Huiya Li
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Pinyi Liu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Bing Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Zengqiang Yuan
- The Brain Science Centre, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- Centre of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Mengdi Guo
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Xinxin Zou
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Yi Qian
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Shiji Deng
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Liwen Zhu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Tao Tao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Shengnan Xia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Xinyu Bao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China.
- Nanjing Neurology Medical Centre, Nanjing, 210008, China.
| |
Collapse
|
8
|
Kashiwabara L, Pirard L, Debier C, Crocker D, Khudyakov J. Effects of cortisol, epinephrine, and bisphenol contaminants on the transcriptional landscape of marine mammal blubber. Am J Physiol Regul Integr Comp Physiol 2023; 325:R504-R522. [PMID: 37602383 DOI: 10.1152/ajpregu.00165.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/04/2023] [Accepted: 08/16/2023] [Indexed: 08/22/2023]
Abstract
Top ocean predators such as marine mammals are threatened by intensifying anthropogenic activity, and understanding the combined effects of multiple stressors on their physiology is critical for conservation efforts. We investigated potential interactions between stress hormones and bisphenol contaminants in a model marine mammal, the northern elephant seal (NES). We exposed precision-cut adipose tissue slices (PCATS) from blubber of weaned NES pups to cortisol (CORT), epinephrine (EPI), bisphenol A (BPA), bisphenol S (BPS), or their combinations (CORT-EPI, BPA-EPI, and BPS-EPI) ex vivo and identified hundreds of genes that were differentially regulated in response to these treatments. CORT altered expression of genes associated with lipolysis and adipogenesis, whereas EPI and CORT-EPI-regulated genes were associated with responses to hormones, lipid and protein turnover, immune function, and transcriptional and epigenetic regulation of gene expression, suggesting that EPI has wide-ranging and prolonged impacts on the transcriptional landscape and function of blubber. Bisphenol treatments alone had a weak impact on gene expression compared with stress hormones. However, the combination of EPI with bisphenols altered expression of genes associated with inflammation, cell stress, DNA damage, regulation of nuclear hormone receptor activity, cell cycle, mitochondrial function, primary ciliogenesis, and lipid metabolism in blubber. Our results suggest that CORT, EPI, bisphenols, and their combinations impact cellular, immune, and metabolic homeostasis in marine mammal blubber, which may affect the ability of marine mammals to sustain prolonged fasting during reproduction and migration, renew tissues, and mount appropriate responses to immune challenges and additional stressors.
Collapse
Affiliation(s)
- Lauren Kashiwabara
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States
| | - Laura Pirard
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la Neuve, Belgium
| | - Cathy Debier
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la Neuve, Belgium
| | - Daniel Crocker
- Department of Biology, Sonoma State University, Rohnert Park, California, United States
| | - Jane Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States
| |
Collapse
|
9
|
Correia CM, Præstholm SM, Havelund JF, Pedersen FB, Siersbæk MS, Ebbesen MF, Gerhart-Hines Z, Heeren J, Brewer J, Larsen S, Blagoev B, Færgeman NJ, Grøntved L. Acute Deletion of the Glucocorticoid Receptor in Hepatocytes Disrupts Postprandial Lipid Metabolism in Male Mice. Endocrinology 2023; 164:bqad128. [PMID: 37610219 DOI: 10.1210/endocr/bqad128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/09/2023] [Accepted: 08/21/2023] [Indexed: 08/24/2023]
Abstract
Hepatic lipid metabolism is highly dynamic, and disruption of several circadian transcriptional regulators results in hepatic steatosis. This includes genetic disruption of the glucocorticoid receptor (GR) as the liver develops. To address the functional role of GR in the adult liver, we used an acute hepatocyte-specific GR knockout model to study temporal hepatic lipid metabolism governed by GR at several preprandial and postprandial circadian timepoints. Lipidomics analysis revealed significant temporal lipid metabolism, where GR disruption results in impaired regulation of specific triglycerides, nonesterified fatty acids, and sphingolipids. This correlates with increased number and size of lipid droplets and mildly reduced mitochondrial respiration, most noticeably in the postprandial phase. Proteomics and transcriptomics analyses suggest that dysregulated lipid metabolism originates from pronounced induced expression of enzymes involved in fatty acid synthesis, β-oxidation, and sphingolipid metabolism. Integration of GR cistromic data suggests that induced gene expression is a result of regulatory actions secondary to direct GR effects on gene transcription.
Collapse
Affiliation(s)
- Catarina Mendes Correia
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Stine Marie Præstholm
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Jesper Foged Havelund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Felix Boel Pedersen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Majken Storm Siersbæk
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Morten Frendø Ebbesen
- DaMBIC, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Zach Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research (CBMR), Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonathan Brewer
- DaMBIC, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Steen Larsen
- Xlab, Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Nils Joakim Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| |
Collapse
|
10
|
Torres N, Tobón-Cornejo S, Velazquez-Villegas LA, Noriega LG, Alemán-Escondrillas G, Tovar AR. Amino Acid Catabolism: An Overlooked Area of Metabolism. Nutrients 2023; 15:3378. [PMID: 37571315 PMCID: PMC10421169 DOI: 10.3390/nu15153378] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Amino acids have been extensively studied in nutrition, mainly as key elements for maintaining optimal protein synthesis in the body as well as precursors of various nitrogen-containing compounds. However, it is now known that amino acid catabolism is an important element for the metabolic control of different biological processes, although it is still a developing field to have a deeper understanding of its biological implications. The mechanisms involved in the regulation of amino acid catabolism now include the contribution of the gut microbiota to amino acid oxidation and metabolite generation in the intestine, the molecular mechanisms of transcriptional control, and the participation of specific miRNAs involved in the regulation of amino acid degrading enzymes. In addition, molecules derived from amino acid catabolism play a role in metabolism as they are used in the epigenetic regulation of many genes. Thus, this review aims to examine the mechanisms of amino acid catabolism and to support the idea that this process is associated with the immune response, abnormalities during obesity, in particular insulin resistance, and the regulation of thermogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Armando R. Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No 15. Col Belisario Domínguez-Sección XVI, Tlalpan, Mexico City 14080, Mexico; (N.T.); (S.T.-C.); (L.A.V.-V.); (L.G.N.); (G.A.-E.)
| |
Collapse
|
11
|
Liu N, Yang X, Guo J, Zhang L, Huang S, Chen J, Huang J, Chen Y, Cui T, Zheng Y, Li T, Tang K, Zhong Y, Duan S, Yu L, Tang Y, Zheng D, Pan H, Gao Y. Hepatic ZBTB22 promotes hyperglycemia and insulin resistance via PEPCK1-driven gluconeogenesis. EMBO Rep 2023; 24:e56390. [PMID: 37154299 PMCID: PMC10240208 DOI: 10.15252/embr.202256390] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/31/2023] [Accepted: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
Excessive gluconeogenesis can lead to hyperglycemia and diabetes through as yet incompletely understood mechanisms. Herein, we show that hepatic ZBTB22 expression is increased in both diabetic clinical samples and mice, being affected by nutritional status and hormones. Hepatic ZBTB22 overexpression increases the expression of gluconeogenic and lipogenic genes, heightening glucose output and lipids accumulation in mouse primary hepatocytes (MPHs), while ZBTB22 knockdown elicits opposite effects. Hepatic ZBTB22 overexpression induces glucose intolerance and insulin resistance, accompanied by moderate hepatosteatosis, while ZBTB22-deficient mice display improved energy expenditure, glucose tolerance, and insulin sensitivity, and reduced hepatic steatosis. Moreover, hepatic ZBTB22 knockout beneficially regulates gluconeogenic and lipogenic genes, thereby alleviating glucose intolerance, insulin resistance, and liver steatosis in db/db mice. ZBTB22 directly binds to the promoter region of PCK1 to enhance its expression and increase gluconeogenesis. PCK1 silencing markedly abolishes the effects of ZBTB22 overexpression on glucose and lipid metabolism in both MPHs and mice, along with the corresponding changes in gene expression. In conclusion, targeting hepatic ZBTB22/PEPCK1 provides a potential therapeutic approach for diabetes.
Collapse
Affiliation(s)
- Naihua Liu
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
- Key Specialty of Clinical PharmacyThe First Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhouChina
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorNanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| | - Jingyi Guo
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Lei Zhang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Shangyi Huang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Jiabing Chen
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Jiawen Huang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yingjian Chen
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Tianqi Cui
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yi Zheng
- Faculty of Chinese MedicineMacau University of Science and TechnologyMacauChina
| | - Tianyao Li
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Kaijia Tang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yadi Zhong
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Siwei Duan
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Lili Yu
- Faculty of Chinese MedicineMacau University of Science and TechnologyMacauChina
| | - Ying Tang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorNanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
| | - Dayong Zheng
- Department of HepatologyTCM‐Integrated Hospital of Southern Medical UniversityGuangzhouChina
- Department of HepatopancreatobiliaryCancer Center, Southern Medical UniversityGuangzhouChina
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhou University of Chinese MedicineGuangzhouChina
| | - Huafeng Pan
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorNanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
| | - Yong Gao
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorNanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
- Division of Hypothalamic Research, Department of Internal MedicineThe University of Texas Southwestern Medical Center at DallasTXDallasUSA
| |
Collapse
|
12
|
Holeček M. Roles of malate and aspartate in gluconeogenesis in various physiological and pathological states. Metabolism 2023:155614. [PMID: 37286128 DOI: 10.1016/j.metabol.2023.155614] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/09/2023]
Abstract
Gluconeogenesis, a pathway for glucose synthesis from non-carbohydrate substances, begins with the synthesis of oxaloacetate (OA) from pyruvate and intermediates of citric acid cycle in hepatocyte mitochondria. The traditional view is that OA does not cross the mitochondrial membrane and must be shuttled to the cytosol, where most enzymes involved in gluconeogenesis are compartmentalized, in the form of malate. Thus, the possibility of transporting OA in the form of aspartate has been ignored. In the article is shown that malate supply to the cytosol increases only when fatty acid oxidation in the liver is activated, such as during starvation or untreated diabetes. Alternatively, aspartate synthesized from OA by mitochondrial aspartate aminotransferase (AST) is transported to the cytosol in exchange for glutamate via the aspartate-glutamate carrier 2 (AGC2). If the main substrate for gluconeogenesis is an amino acid, aspartate is converted to OA via urea cycle, therefore, ammonia detoxification and gluconeogenesis are simultaneously activated. If the main substrate is lactate, OA is synthesized by cytosolic AST, glutamate is transported to the mitochondria through AGC2, and nitrogen is not lost. It is concluded that, compared to malate, aspartate is a more suitable form of OA transport from the mitochondria for gluconeogenesis.
Collapse
Affiliation(s)
- Milan Holeček
- Department of Physiology, Charles University, Faculty of Medicine in Hradec Králové, Czech Republic.
| |
Collapse
|
13
|
Tao Y, Jiang Q, Wang Q. Adipose tissue macrophages in remote modulation of hepatic glucose production. Front Immunol 2022; 13:998947. [PMID: 36091076 PMCID: PMC9449693 DOI: 10.3389/fimmu.2022.998947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatic glucose production (HGP) is fine-regulated via glycogenolysis or gluconeogenesis to maintain physiological concentration of blood glucose during fasting-feeding cycle. Aberrant HGP leads to hyperglycemia in obesity-associated diabetes. Adipose tissue cooperates with the liver to regulate glycolipid metabolism. During these processes, adipose tissue macrophages (ATMs) change their profiles with various physio-pathological settings, producing diverse effects on HGP. Here, we briefly review the distinct phenotypes of ATMs under different nutrition states including feeding, fasting or overnutrition, and detail their effects on HGP. We discuss several pathways by which ATMs regulate hepatic gluconeogenesis or glycogenolysis, leading to favorable or unfavorable metabolic consequences. Furthermore, we summarize emerging therapeutic targets to correct metabolic disorders in morbid obesity or diabetes based on ATM-HGP axis. This review puts forward the importance and flexibility of ATMs in regulating HGP, proposing ATM-based HGP modulation as a potential therapeutic approach for obesity-associated metabolic dysfunction.
Collapse
Affiliation(s)
| | | | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
14
|
Cant J, Reyes G, Seymour D. Review: Influence of postabsorptive metabolism on essential amino acid partitioning in lactating dairy cows. Animal 2022; 16 Suppl 3:100573. [DOI: 10.1016/j.animal.2022.100573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/24/2022] [Accepted: 05/30/2022] [Indexed: 01/07/2023] Open
|
15
|
Goldberg D, Charni-Natan M, Buchshtab N, Bar-Shimon M, Goldstein I. Hormone-controlled cooperative binding of transcription factors drives synergistic induction of fasting-regulated genes. Nucleic Acids Res 2022; 50:5528-5544. [PMID: 35556130 PMCID: PMC9177981 DOI: 10.1093/nar/gkac358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/22/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
During fasting, hepatocytes produce glucose in response to hormonal signals. Glucagon and glucocorticoids are principal fasting hormones that cooperate in regulating glucose production via gluconeogenesis. However, how these hormone signals are integrated and interpreted to a biological output is unknown. Here, we use genome-wide profiling of gene expression, enhancer dynamics and transcription factor (TF) binding in primary mouse hepatocytes to uncover the mode of cooperation between glucagon and glucocorticoids. We found that compared to a single treatment with each hormone, a dual treatment directs hepatocytes to a pro-gluconeogenic gene program by synergistically inducing gluconeogenic genes. The cooperative mechanism driving synergistic gene expression is based on ‘assisted loading’ whereby a glucagon-activated TF (cAMP responsive element binding protein; CREB) leads to enhancer activation which facilitates binding of the glucocorticoid receptor (GR) upon glucocorticoid stimulation. Glucagon does not only activate single enhancers but also activates enhancer clusters, thereby assisting the loading of GR also across enhancer units within the cluster. In summary, we show that cells integrate extracellular signals by an enhancer-specific mechanism: one hormone-activated TF activates enhancers, thereby assisting the loading of a TF stimulated by a second hormone, leading to synergistic gene induction and a tailored transcriptional response to fasting.
Collapse
Affiliation(s)
- Dana Goldberg
- Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food and Environment. The Hebrew University of Jerusalem. POB 12, Rehovot 7610001, Israel
| | - Meital Charni-Natan
- Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food and Environment. The Hebrew University of Jerusalem. POB 12, Rehovot 7610001, Israel
| | - Nufar Buchshtab
- Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food and Environment. The Hebrew University of Jerusalem. POB 12, Rehovot 7610001, Israel
| | - Meirav Bar-Shimon
- Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food and Environment. The Hebrew University of Jerusalem. POB 12, Rehovot 7610001, Israel
| | - Ido Goldstein
- Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food and Environment. The Hebrew University of Jerusalem. POB 12, Rehovot 7610001, Israel
| |
Collapse
|
16
|
Arora P, Singh K, Kumari M, Trivedi R. Temporal profile of serum metabolites and inflammation following closed head injury in rats is associated with HPA axis hyperactivity. Metabolomics 2022; 18:28. [PMID: 35486220 DOI: 10.1007/s11306-022-01886-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Closed head injury (CHI) causes neurological disability along with systemic alterations that can activate neuro-endocrine response through hypothalamic-pituitary-adrenal (HPA) axis activation. A dysregulated HPA axis function can lead to relocation of energy substrates and alteration in metabolic pathways and inflammation at the systemic level. OBJECTIVES Assessment of time-dependent changes in serum metabolites and inflammation after both mild and moderate CHI. Along with this, serum corticosterone levels and hypothalamic microglial response were observed. METHODS Rats underwent mild and moderate weight-drop injury and their serum and hypothalamus were assessed at acute, sub-acute and chronic timepoints. Changes in serum metabolomics were determined using high resolution NMR spectroscopy. Serum inflammatory cytokine, corticosterone levels and hypothalamic microglia were assessed at all timepoints. RESULTS Metabolites including lactate, choline and branched chain amino acids were found as the classifiers that helped distinguish between control and injured rats during acute, sub-acute and chronic timepoints. While, increased αglucose: βglucose and TMAO: choline ratios after acute and sub-acute timepoints of mild injury differentiated from moderate injured rats. The injured rats also showed distinct inflammatory profile where IL-1β and TNF-α levels were upregulated in moderate injured rats while IL-10 levels were downregulated in mild injured rats. Furthermore, injury specific alterations in serum metabolic and immunologic profile were found to be associated with hyperactive HPA axis, with consistent increase in serum corticosterone concentration post injury. The hypothalamic microglia showed a characteristic activated de-ramified cellular morphology in both mild and moderate injured rats. CONCLUSION The study suggests that HPA axis hyperactivity along with hypothalamic microglial activation led to temporal changes in the systemic metabolism and inflammation. These time dependent changes in the metabolite profile of rats can further strengthen the knowledge of diagnostic markers and help distinguish injury related outcomes after TBI.
Collapse
Affiliation(s)
- Palkin Arora
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, 110054, India
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Kavita Singh
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, 110054, India
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Megha Kumari
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, 110054, India
- Department of Biotechnology, Delhi Technological University (DTU), Delhi, 110042, India
| | - Richa Trivedi
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, 110054, India.
| |
Collapse
|
17
|
Zmazek J, Grubelnik V, Markovič R, Marhl M. Modeling the Amino Acid Effect on Glucagon Secretion from Pancreatic Alpha Cells. Metabolites 2022; 12:metabo12040348. [PMID: 35448534 PMCID: PMC9028923 DOI: 10.3390/metabo12040348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) is a burdensome problem in modern society, and intensive research is focused on better understanding the underlying cellular mechanisms of hormone secretion for blood glucose regulation. T2DM is a bi-hormonal disease, and in addition to 100 years of increasing knowledge about the importance of insulin, the second hormone glucagon, secreted by pancreatic alpha cells, is becoming increasingly important. We have developed a mathematical model for glucagon secretion that incorporates all major metabolic processes of glucose, fatty acids, and glutamine as the most abundant postprandial amino acid in blood. In addition, we consider cAMP signaling in alpha cells. The model predictions quantitatively estimate the relative importance of specific metabolic and signaling pathways and particularly emphasize the important role of glutamine in promoting glucagon secretion, which is in good agreement with known experimental data.
Collapse
Affiliation(s)
- Jan Zmazek
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia; (J.Z.); (R.M.)
| | - Vladimir Grubelnik
- Faculty of Electrical Engineering and Computer Science, University of Maribor, 2000 Maribor, Slovenia;
| | - Rene Markovič
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia; (J.Z.); (R.M.)
- Faculty of Electrical Engineering and Computer Science, University of Maribor, 2000 Maribor, Slovenia;
| | - Marko Marhl
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia; (J.Z.); (R.M.)
- Faculty of Education, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Correspondence:
| |
Collapse
|
18
|
Lu Y, Shi C, Jin X, He J, Yin Z. Domestication of farmed fish via the attenuation of stress responses mediated by the hypothalamus-pituitary-inter-renal endocrine axis. Front Endocrinol (Lausanne) 2022; 13:923475. [PMID: 35937837 PMCID: PMC9353172 DOI: 10.3389/fendo.2022.923475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/29/2022] [Indexed: 12/13/2022] Open
Abstract
Human-directed domestication of terrestrial animals traditionally requires thousands of years for breeding. The most prominent behavioral features of domesticated animals include reduced aggression and enhanced tameness relative to their wild forebears, and such behaviors improve the social tolerance of domestic animals toward both humans and crowds of their own species. These behavioral responses are primarily mediated by the hypothalamic-pituitary-adrenal (inter-renal in fish) (HPA/I) endocrine axis, which is involved in the rapid conversion of neuronal-derived perceptual information into hormonal signals. Over recent decades, growing evidence implicating the attenuation of the HPA/I axis during the domestication of animals have been identified through comprehensive genomic analyses of the paleogenomic datasets of wild progenitors and their domestic congeners. Compared with that of terrestrial animals, domestication of most farmed fish species remains at early stages. The present review focuses on the application of HPI signaling attenuation to accelerate the domestication and genetic breeding of farmed fish. We anticipate that deeper understanding of HPI signaling and its implementation in the domestication of farmed fish will benefit genetic breeding to meet the global demands of the aquaculture industry.
Collapse
Affiliation(s)
- Yao Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Chuang Shi
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Xia Jin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jiangyan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
- Hubei Hongshan Laboratory, Wuhan, China
- Hainan Yazhou Bay Seed Laboratory, Sanya, China
- *Correspondence: Zhan Yin,
| |
Collapse
|