1
|
Li H, Ren Q, Shi M, Ma L, Fu P. Lactate metabolism and acute kidney injury. Chin Med J (Engl) 2025; 138:916-924. [PMID: 38802283 PMCID: PMC12037090 DOI: 10.1097/cm9.0000000000003142] [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: 01/22/2024] [Indexed: 05/29/2024] Open
Abstract
ABSTRACT Acute kidney injury (AKI) is a common clinically critical syndrome in hospitalized patients with high morbidity and mortality. At present, the mechanism of AKI has not been fully elucidated, and no therapeutic drugs exist. As known, glycolytic product lactate is a key metabolite in physiological and pathological processes. The kidney is an important gluconeogenic organ, where lactate is the primary substrate of renal gluconeogenesis in physiological conditions. During AKI, altered glycolysis and gluconeogenesis in kidneys significantly disturb the lactate metabolic balance, which exert impacts on the severity and prognosis of AKI. Additionally, lactate-derived posttranslational modification, namely lactylation, is novel to AKI as it could regulate gene transcription of metabolic enzymes involved in glycolysis or Warburg effect. Protein lactylation widely exists in human tissues and may severely affect non-histone functions. Moreover, the strategies of intervening lactate metabolic pathways are expected to bring a new dawn for the treatment of AKI. This review focused on renal lactate metabolism, especially in proximal renal tubules after AKI, and updated recent advances of lactylation modification, which may help to explore potential therapeutic targets against AKI.
Collapse
Affiliation(s)
- Hui Li
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Qian Ren
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Min Shi
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Ma
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Fu
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
2
|
Rahim M, Bednarski TK, Hasenour CM, Banerjee DR, Trenary I, Young JD. Simultaneous in vivo multi-organ fluxomics reveals divergent metabolic adaptations in liver, heart, and skeletal muscle during obesity. Cell Rep 2025; 44:115591. [PMID: 40244853 DOI: 10.1016/j.celrep.2025.115591] [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: 02/03/2024] [Revised: 02/23/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
We present an isotope-based metabolic flux analysis (MFA) approach to simultaneously quantify metabolic fluxes in the liver, heart, and skeletal muscle of individual mice. The platform was scaled to examine metabolic flux adaptations in age-matched cohorts of mice exhibiting varying levels of chronic obesity. We found that severe obesity increases hepatic gluconeogenesis and citric acid cycle flux, accompanied by elevated glucose oxidation in the heart that compensates for impaired fatty acid oxidation. In contrast, skeletal muscle fluxes exhibit an overall reduction in substrate oxidation. These findings demonstrate the dichotomy in fuel utilization between cardiac and skeletal muscle during worsening metabolic disease and demonstrate the divergent effects of obesity on metabolic fluxes in different organs. This multi-tissue MFA technology can be extended to address important questions about in vivo regulation of metabolism and its dysregulation in disease, which cannot be fully answered through studies of single organs or isolated cells/tissues.
Collapse
Affiliation(s)
- Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Tomasz K Bednarski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Clinton M Hasenour
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Deveena R Banerjee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Irina Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
3
|
Ma X, Li B, Liu Y, Guo X. An inverted U-shaped association between high-sensitivity C-reactive protein and the albumin ratio and hepatic steatosis and liver fibrosis: a population-based study. Front Nutr 2025; 12:1534200. [PMID: 40303878 PMCID: PMC12037389 DOI: 10.3389/fnut.2025.1534200] [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: 11/28/2024] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Background The high-sensitivity C-reactive protein to albumin (CAR) ratio is a comprehensive measure of inflammation in vivo. Hepatic steatosis and fibrosis are significantly correlated with inflammation. The present study aimed to explore the possible associations between CAR and hepatic steatosis and fibrosis in the American population. Methods The study population involved the National Health and Nutrition Examination Survey (NHANES) participants from 2017 to 2020. The natural logarithm of CAR, calculated as Ln(CAR) with base "e," was used for further analyses. The relationships between Ln(CAR) and the controlled attenuation parameter (CAP) and between Ln(CAR) and liver stiffness measurement (LSM) were investigated through multivariate linear regression analysis. Interaction and subgroup analysis identified factors affecting these variables. Nonlinear relationships were elucidated by smoothing curves and threshold effect analysis. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the predictive performance of the CAR for non-alcoholic fatty liver disease (NAFLD). The results were adjusted for U.S. population estimates. Results The study included a total of 7,404 individuals. Ln(CAR) was positively correlated with CAP in the fully adjusted model, with an effect value of β = 1.827 (95% CI, 0.611, 3.042). A more pronounced positive association was observed among participants with a BMI ≥ 25 kg/m2 in the subgroup analysis. An inverted U-shaped association was shown between Ln(CAR) and CAP through smooth curve fitting and a two-segment linear regression model, with an inflection point of (-9.594). ROC curve analysis showed that CAR had a moderate predictive value for NAFLD (AUC = 0.6895), with a sensitivity of 0.7276 and a specificity of 0.6092. No significant association was detected between Ln(CAR) and the LSM. Conclusion We demonstrate an inverted U-shaped relationship between Ln(CAR) and CAP risk within the U.S. demographic. Our results suggest that CAR may serve as a valuable diagnostic tool for NAFLD. Further prospective research is necessary to validate this conclusion.
Collapse
Affiliation(s)
| | | | | | - Xiaoyan Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
4
|
Huang H, Han Y, Zhang Y, Zeng J, He X, Cheng J, Wang S, Xiong Y, Yin H, Yuan Q, Huang L, Xie Y, Meng J, Tao L, Peng Z. Deletion of Pyruvate Carboxylase in Tubular Epithelial Cell Promotes Renal Fibrosis by Regulating SQOR/cGAS/STING-Mediated Glycolysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408753. [PMID: 39836535 PMCID: PMC11967762 DOI: 10.1002/advs.202408753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 12/31/2024] [Indexed: 01/23/2025]
Abstract
Renal fibrosis is a common pathway involved in the progression of various chronic kidney diseases to end-stage renal disease. Recent studies show that mitochondrial injury of renal tubular epithelial cells (RTECs) is a crucial pathological foundation for renal fibrosis. However, the underlying regulatory mechanisms remain unclear. Pyruvate carboxylase (PC) is a catalytic enzyme located within the mitochondria that is intricately linked with mitochondrial damage and metabolism. In the present study, the downregulation of PC in various fibrotic animal and human kidney samples is demonstrated. Renal proximal tubule-specific Pcx gene knockout mice (PcxcKO) has significant interstitial fibrosis compared to control mice, with heightened expression of extracellular matrix molecules. This is further demonstrated in a stable PC knock-out RTEC line. Mechanistically, PC deficiency reduces its interaction with sulfide:quinone oxidoreductase (SQOR), increasing the ubiquitination and degradation of SQOR. This leads to mitochondrial morphological and functional disruption, increased mtDNA release, activation of the cGAS-STING pathway, and elevated glycolysis levels, and ultimately, promotes renal fibrosis. This study investigates the molecular mechanisms through which PC deficiency induces mitochondrial injury and metabolic reprogramming in RTECs. This study provides a novel theoretical foundation and potential therapeutic targets for the pathogenesis and treatment of renal fibrosis.
Collapse
Affiliation(s)
- Hao Huang
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Department of Cell biologySchool of Life SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
| | - Yuanyuan Han
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Yan Zhang
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Jianhua Zeng
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Xin He
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Jiawei Cheng
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Songkai Wang
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Yiwei Xiong
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Hongling Yin
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- Department of Pathology, Xiangya HospitalCentral South UniversityChangsha410008China
| | - Qiongjing Yuan
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Ling Huang
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Yanyun Xie
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Jie Meng
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- Department of Pulmonary and Critical Care Medicine, Third Xiangya HospitalCentral South UniversityChangsha410013China
| | - Lijian Tao
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| |
Collapse
|
5
|
Park JM, Lin SH, Baxter JD, Harrison CE, Leary J, Mozingo C, Liticker J, Malloy CR, Jin ES. Disrupted metabolic flux balance between pyruvate dehydrogenase and pyruvate carboxylase in human fatty liver. Metabolism 2025; 165:156151. [PMID: 39890055 PMCID: PMC11955189 DOI: 10.1016/j.metabol.2025.156151] [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: 11/27/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Hepatic metabolism involving pyruvate carboxylase (PC) and pyruvate dehydrogenase (PDH) may be abnormal in fatty livers. In this study, [13C]bicarbonate production from [1-13C1]pyruvate in the liver and glycerol glyceroneogenesis were examined in relation to hepatic fat content using hyperpolarized [1-13C1]pyruvate and oral [U-13C3]glycerol. After an overnight fast, 15 subjects with a range of hepatic fat content received hyperpolarized [1-13C1]pyruvate intravenously to assess its conversion to [1-13C1]lactate and [13C]bicarbonate in the liver. They also received oral [U-13C3]glycerol, followed by venous blood sampling to examine glucose and the glycerol backbone of the triglycerides released primarily from the liver. From hyperpolarized [1-13C1]pyruvate, participants with high intrahepatic fat fraction produced higher [1-13C1]lactate and lower [13C]bicarbonate than those with low liver fat. The fraction of plasma triglycerides derived from oral [U-13C3]glycerol via the TCA cycle was similar between groups. The fraction of plasma [5,6-13C2]glucose, which reflects PC flux, decreased in subjects with fatty liver. In contrast, the fraction of [4,5-13C2]glucose + [6-13C1]glucose, which can be produced via either PC or PDH, was comparable between groups. The study results suggest a shift in pyruvate metabolism in fatty liver, with a decreased metabolic flux ratio of PC/PDH. The methodology in this study provides insights into fatty liver metabolism of human subjects inaccessible previously and is applicable to advanced liver diseases such as cirrhosis and hepatomas.
Collapse
Affiliation(s)
- Jae Mo Park
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA; Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA; Department of Radiology, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA.
| | - Sung-Han Lin
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA
| | - Jeannie D Baxter
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA
| | - Crystal E Harrison
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA
| | - Jennine Leary
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA
| | - Corey Mozingo
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA
| | - Jeff Liticker
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA
| | - Craig R Malloy
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA; Department of Radiology, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA; Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA; VA North Texas Healthcare System, Dallas 75216, TX, USA.
| | - Eunsook S Jin
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA; Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas 75390, TX, USA.
| |
Collapse
|
6
|
Yuan B, Doxsey W, Tok Ö, Kwon YY, Liang Y, Inouye KE, Hotamışlıgil GS, Hui S. An organism-level quantitative flux model of energy metabolism in mice. Cell Metab 2025; 37:1012-1023.e6. [PMID: 39983714 PMCID: PMC11964847 DOI: 10.1016/j.cmet.2025.01.008] [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: 02/26/2024] [Revised: 11/21/2024] [Accepted: 01/09/2025] [Indexed: 02/23/2025]
Abstract
Mammalian tissues feed on nutrients in the blood circulation. At the organism level, mammalian energy metabolism is comprised of the oxidation, storage, interconversion, and release of circulating nutrients. Here, by integrating isotope tracer infusion, mass spectrometry, and isotope gas analyzer measurement, we developed a framework to systematically quantify fluxes through these metabolic processes for 10 major circulating energy nutrients in mice, resulting in an organism-level quantitative flux model of energy metabolism. This model revealed in wild-type mice that circulating nutrients have metabolic cycling fluxes dominant to their oxidation fluxes, with distinct partitions between cycling and oxidation for individual circulating nutrients. Applications of this framework in obese mouse models showed extensive elevation of metabolic cycling fluxes in ob/ob mice but not in diet-induced obese mice on a per-animal or per-lean mass basis. Our framework is a valuable tool to reveal new features of energy metabolism in physiological and disease conditions.
Collapse
Affiliation(s)
- Bo Yuan
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Will Doxsey
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Özlem Tok
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Young-Yon Kwon
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Yanshan Liang
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Karen E Inouye
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gökhan S Hotamışlıgil
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Sheng Hui
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
7
|
Zhou Y, Que T, Yu L, Que S, Xu J, Liu Z. Current understanding on inferior quality of liver grafts by donation after circulatory death based on multi-omics data. Front Immunol 2025; 16:1548735. [PMID: 40181961 PMCID: PMC11965662 DOI: 10.3389/fimmu.2025.1548735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Given the inevitable hypoxia and reperfusion injury that occur in organs donated after circulatory death (DCD), the quality and function of these organs are significantly compromised, greatly limiting their application in clinical organ transplantation. Recently, the advancement of functional omics technologies has enabled us to deeply analyze the mechanisms underlying DCD donor organ damage from multiple perspectives. This review systematically integrates the studies from transcriptomics, proteomics, and metabolomics to reveal the key biological mechanisms associated with the declines in DCD donor organ quality, including oxidative stress, inflammatory responses, cell death pathways, and metabolic disturbances. Additionally, we summarized emerging therapeutic strategies based on findings from omics perspectives, offering new possibilities to improve the quality of DCD organ for better transplant prognosis. Finally, we discussed the challenges in current research and future directions to provide scientific evidence for clinical practice and promote the application of DCD donors in organ transplantation.
Collapse
Affiliation(s)
- Yifeng Zhou
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Ting Que
- Birth Defects Prevention and Control Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lu Yu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Shulan (Hangzhou) Hospital, Hangzhou, China
| | | | - Jun Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, School of Medicine, Chinese Academy of Medical Sciences, First Affiliated Hospital, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhengtao Liu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- Shulan (Hangzhou) Hospital, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, School of Medicine, Chinese Academy of Medical Sciences, First Affiliated Hospital, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Saito T, Espe M, Mommens M, Bock C, Fernandes JM, Skjærven KH. Altered spawning seasons of Atlantic salmon broodstock transcriptionally and epigenetically influence cell cycle and lipid-mediated regulations in their offspring. PLoS One 2025; 20:e0317770. [PMID: 39992963 PMCID: PMC11849821 DOI: 10.1371/journal.pone.0317770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/03/2025] [Indexed: 02/26/2025] Open
Abstract
Manipulating spawning seasons of Atlantic salmon (Salmo salar) is a common practice to facilitate year-round harvesting in salmon aquaculture. This process involves adjusting water temperature and light regime to control female broodstock maturation. However, recent studies have indicated that altered spawning seasons can significantly affect the nutritional status and growth performance of the offspring. Therefore, gaining a deeper understanding of the biological regulations influenced by these alterations is crucial to enhance the growth performance of fish over multiple generations. In this study, we investigated omics data from four different spawning seasons achieved through recirculating aquaculture systems (RAS) and sea-pen-based approaches. In addition to the normal spawning season in November (sea-pen), three altered seasons were designated: off-season (five-month advance, RAS), early season (two-month advance, sea-pen), and late season (two-month delay, sea-pen). We conducted comprehensive gene expression and DNA methylation analysis on liver samples collected from the start-feeding larvae of the next generation. Our results revealed distinct gene expression and DNA methylation patterns associated with the altered spawning seasons. Specifically, offspring from RAS-based off-season exhibited altered lipid-mediated regulation, while those from sea-pen-based early and late seasons showed changes in cellular processes, particularly in cell cycle regulation when compared to the normal season. The consequences of our findings are significant for growth and health, potentially providing information for developing valuable tools for assessing growth potential and optimizing production strategies in aquaculture.
Collapse
Affiliation(s)
| | - Marit Espe
- Institute of Marine Research, Bergen, Norway
| | | | - Christoph Bock
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | | |
Collapse
|
9
|
Politte H, Maram L, Elgendy B. Advances in the Development of Mitochondrial Pyruvate Carrier Inhibitors for Therapeutic Applications. Biomolecules 2025; 15:223. [PMID: 40001526 PMCID: PMC11852594 DOI: 10.3390/biom15020223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
The mitochondrial pyruvate carrier (MPC) is a transmembrane protein complex critical for cellular energy metabolism, enabling the transport of pyruvate from the cytosol into the mitochondria, where it fuels the citric acid cycle. By regulating this essential entry point of carbon into mitochondrial metabolism, MPC is pivotal for maintaining cellular energy balance and metabolic flexibility. Dysregulation of MPC activity has been implicated in several metabolic disorders, including type 2 diabetes, obesity, and cancer, underscoring its potential as a therapeutic target. This review provides an overview of the MPC complex, examining its structural components, regulatory mechanisms, and biological functions. We explore the current understanding of transcriptional, translational, and post-translational modifications that modulate MPC function and highlight the clinical relevance of MPC dysfunction in metabolic and neurodegenerative diseases. Progress in the development of MPC-targeting therapeutics is discussed, with a focus on challenges in designing selective and potent inhibitors. Emphasis is placed on modern approaches for identifying novel inhibitors, particularly virtual screening and computational strategies. This review establishes a foundation for further research into the medicinal chemistry of MPC inhibitors, promoting advances in structure-based drug design to develop therapeutics for metabolic and neurodegenerative diseases.
Collapse
Affiliation(s)
- Henry Politte
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; (H.P.); (L.M.)
- Center for Clinical Pharmacology, Washington University School of Medicine and University of Health Sciences and Pharmacy, St. Louis, MO 63110, USA
| | - Lingaiah Maram
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; (H.P.); (L.M.)
- Center for Clinical Pharmacology, Washington University School of Medicine and University of Health Sciences and Pharmacy, St. Louis, MO 63110, USA
| | - Bahaa Elgendy
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; (H.P.); (L.M.)
- Center for Clinical Pharmacology, Washington University School of Medicine and University of Health Sciences and Pharmacy, St. Louis, MO 63110, USA
| |
Collapse
|
10
|
Rauckhorst AJ, Sheldon RD, Pape DJ, Ahmed A, Falls-Hubert KC, Merrill RA, Brown RF, Deshmukh K, Vallim TA, Deja S, Burgess SC, Taylor EB. A hierarchical hepatic de novo lipogenesis substrate supply network utilizing pyruvate, acetate, and ketones. Cell Metab 2025; 37:255-273.e6. [PMID: 39471817 PMCID: PMC11856365 DOI: 10.1016/j.cmet.2024.10.013] [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: 11/03/2023] [Revised: 08/07/2024] [Accepted: 10/18/2024] [Indexed: 11/01/2024]
Abstract
Hepatic de novo lipogenesis (DNL) is a fundamental physiologic process that is often pathogenically elevated in metabolic disease. Treatment is limited by incomplete understanding of the metabolic pathways supplying cytosolic acetyl-CoA, the obligate precursor to DNL, including their interactions and proportional contributions. Here, we combined extensive 13C tracing with liver-specific knockout of key mitochondrial and cytosolic proteins mediating cytosolic acetyl-CoA production. We show that the mitochondrial pyruvate carrier (MPC) and ATP-citrate lyase (ACLY) gate the major hepatic lipogenic acetyl-CoA production pathway, operating in parallel with acetyl-CoA synthetase 2 (ACSS2). Given persistent DNL after mitochondrial citrate carrier (CiC) and ACSS2 double knockout, we tested the contribution of exogenous and leucine-derived acetoacetate to acetoacetyl-CoA synthetase (AACS)-dependent DNL. CiC knockout increased acetoacetate-supplied hepatic acetyl-CoA production and DNL, indicating that ketones function as mitochondrial-citrate reciprocal DNL precursors. By delineating a mitochondrial-cytosolic DNL substrate supply network, these findings may inform strategies to therapeutically modulate DNL.
Collapse
Affiliation(s)
- Adam J Rauckhorst
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; FOEDRC Metabolomics Core Research Facility, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Ryan D Sheldon
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Daniel J Pape
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Adnan Ahmed
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Kelly C Falls-Hubert
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Ronald A Merrill
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Reid F Brown
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; FOEDRC Metabolomics Core Research Facility, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Kshitij Deshmukh
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Thomas A Vallim
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Stanislaw Deja
- Center for Human Nutrition, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Shawn C Burgess
- Center for Human Nutrition, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Eric B Taylor
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; FOEDRC Metabolomics Core Research Facility, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA.
| |
Collapse
|
11
|
Liu JY, Kuna RS, Pinheiro LV, Nguyen PTT, Welles JE, Drummond JM, Murali N, Sharma PV, Supplee JG, Shiue M, Zhao S, Farria AT, Kumar A, Ruchhoeft ML, Demetriadou C, Kantner DS, Chatoff A, Megill E, Titchenell PM, Snyder NW, Metallo CM, Wellen KE. Bempedoic acid suppresses diet-induced hepatic steatosis independently of ATP-citrate lyase. Cell Metab 2025; 37:239-254.e7. [PMID: 39471816 PMCID: PMC11711013 DOI: 10.1016/j.cmet.2024.10.014] [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: 11/03/2023] [Revised: 08/07/2024] [Accepted: 10/18/2024] [Indexed: 11/01/2024]
Abstract
ATP citrate lyase (ACLY) synthesizes acetyl-CoA for de novo lipogenesis (DNL), which is elevated in metabolic dysfunction-associated steatotic liver disease. Hepatic ACLY is inhibited by the LDL-cholesterol-lowering drug bempedoic acid (BPA), which also improves steatosis in mice. While BPA potently suppresses hepatic DNL and increases fat catabolism, it is unclear if ACLY is its primary molecular target in reducing liver triglyceride. We show that on a Western diet, loss of hepatic ACLY alone or together with the acetyl-CoA synthetase ACSS2 unexpectedly exacerbates steatosis, linked to reduced PPARα target gene expression and fatty acid oxidation. Importantly, BPA treatment ameliorates Western diet-mediated triacylglyceride accumulation in both WT and liver ACLY knockout mice, indicating that its primary effects on hepatic steatosis are ACLY independent. Together, these data indicate that hepatic ACLY plays an unexpected role in restraining diet-dependent lipid accumulation and that BPA exerts substantial effects on hepatic lipid metabolism independently of ACLY.
Collapse
Affiliation(s)
- Joyce Y Liu
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ramya S Kuna
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Laura V Pinheiro
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phuong T T Nguyen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jaclyn E Welles
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jack M Drummond
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nivitha Murali
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Prateek V Sharma
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julianna G Supplee
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mia Shiue
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven Zhao
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aimee T Farria
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Avi Kumar
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mauren L Ruchhoeft
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina Demetriadou
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Daniel S Kantner
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Adam Chatoff
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Emily Megill
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Paul M Titchenell
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel W Snyder
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Christian M Metallo
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
Su J, Tian X, Cheng H, Liu D, Wang Z, Sun S, Wang HW, Sui SF. Structural insight into synergistic activation of human 3-methylcrotonyl-CoA carboxylase. Nat Struct Mol Biol 2025; 32:73-85. [PMID: 39223421 DOI: 10.1038/s41594-024-01379-3] [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: 10/21/2023] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
The enzymes 3-methylcrotonyl-coenzyme A (CoA) carboxylase (MCC), pyruvate carboxylase and propionyl-CoA carboxylase belong to the biotin-dependent carboxylase family located in mitochondria. They participate in various metabolic pathways in human such as amino acid metabolism and tricarboxylic acid cycle. Many human diseases are caused by mutations in those enzymes but their structures have not been fully resolved so far. Here we report an optimized purification strategy to obtain high-resolution structures of intact human endogenous MCC, propionyl-CoA carboxylase and pyruvate carboxylase in different conformational states. We also determine the structures of MCC bound to different substrates. Analysis of MCC structures in different states reveals the mechanism of the substrate-induced, multi-element synergistic activation of MCC. These results provide important insights into the catalytic mechanism of the biotin-dependent carboxylase family and are of great value for the development of new drugs for the treatment of related diseases.
Collapse
Affiliation(s)
- Jiayue Su
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structures, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xuyang Tian
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structures, School of Life Sciences, Tsinghua University, Beijing, China
| | - Hang Cheng
- The California Institute for Quantitative Biosciences (QB3), University of California campuses at Berkeley, Berkeley, CA, USA
| | - Desheng Liu
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structures, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ziyi Wang
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structures, School of Life Sciences, Tsinghua University, Beijing, China
| | - Shan Sun
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structures, School of Life Sciences, Tsinghua University, Beijing, China.
| | - Hong-Wei Wang
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structures, School of Life Sciences, Tsinghua University, Beijing, China.
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| | - Sen-Fang Sui
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structures, School of Life Sciences, Tsinghua University, Beijing, China.
- School of Life Sciences, Cryo-EM Center, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
13
|
Li X, Qin X, Xie Y, Wang L, Wang J, Ji S, Jiang H, Wang Q. Metabolic Characterization of Cerebrospinal Fluid for Patients With Autoimmune Encephalitis: A Preliminary Study. CNS Neurosci Ther 2025; 31:e70203. [PMID: 39749658 PMCID: PMC11696248 DOI: 10.1111/cns.70203] [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: 06/09/2024] [Revised: 11/09/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Metabolomics offers promise in uncovering potential biomarkers and understanding the pathophysiology of autoimmune encephalitis (AE), which is a cluster of disorders with the host immune system targeting self-antigens expressed in the central nervous system (CNS). In this research, our objective was to explore metabolic characterization in cerebrospinal fluid (CSF) from individuals with AE, aiming to shed light on the pathophysiology of AE. METHODS A targeted approach was applied using an ultra-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-MS/MS) system to study CSF metabolites in patients with AE (n = 18), and control subjects without neurological diseases (n = 17). RESULTS A total of 21 potential biomarkers were acquired by getting the intersection of the differential metabolites from univariate statistics and multidimensional statistics between the AE (cell-based assay panel, CBA-panel) group and the control group. Specifically, the levels of pyruvic acid and oxoglutaric acid were notably elevated in the AE(CBA-panel) group compared to those in the control group, indicating that the dysregulated TCA cycle may play a pivotal role in the progression of AE(CBA-panel). Interestingly, 27 potential biomarkers were acquired by getting the intersection of the differential metabolites from univariate statistics and multidimensional statistics between the anti-N-methyl-D-aspartate receptor encephalitis (NMDARE) group and the control group, suggesting that the disparities between patients with greater homogeneity and the controls are amplified. In addition, seven differential metabolites were identified by the univariate statistics between the AE (tissue-based assay, TBA) group and the control group, including alpha-linolenic acid and gamma-linolenic acid, suggesting that dysregulated biosynthesis of unsaturated fatty acids and alpha-linolenic acid metabolism might be crucial in the AE(TBA) disease course. CONCLUSION Collectively, distinct metabolic profiles were evident in the CSF of the AE group compared to the control group, notably involving metabolites associated with mitochondrial dysfunction, which helped to elucidate the pathophysiology of AE.
Collapse
Affiliation(s)
- Xiaolong Li
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of Neurology, Xiangyang No. 1 People's HospitalHubei University of MedicineXiangyangHubeiChina
| | - Xiaoxiao Qin
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yuan Xie
- Department of Neurology, Xiangyang No. 1 People's HospitalHubei University of MedicineXiangyangHubeiChina
| | - Lingyun Wang
- Zhangjiang Center for Translational MedicineShanghai Biotecan Pharmaceuticals co. Ltd.ShanghaiChina
| | - Jinwen Wang
- Zhangjiang Center for Translational MedicineShanghai Biotecan Pharmaceuticals co. Ltd.ShanghaiChina
| | - Shushen Ji
- Zhangjiang Center for Translational MedicineShanghai Biotecan Pharmaceuticals co. Ltd.ShanghaiChina
| | - Huihui Jiang
- Zhangjiang Center for Translational MedicineShanghai Biotecan Pharmaceuticals co. Ltd.ShanghaiChina
| | - Qun Wang
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- National Center for Clinical Medicine of Neurological DiseasesBeijingChina
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| |
Collapse
|
14
|
Smith DM, Choi J, Wolfgang MJ. Tissue specific roles of fatty acid oxidation. Adv Biol Regul 2025; 95:101070. [PMID: 39672726 PMCID: PMC11832339 DOI: 10.1016/j.jbior.2024.101070] [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: 11/18/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
Mitochondrial long chain fatty acid β-oxidation is a critical central carbon catabolic process. The importance of fatty acid oxidation is made evident by the life-threatening disease associated with diverse inborn errors in the pathway. While inborn errors show multisystemic requirements for fatty acid oxidation, it is not clear from the clinical presentation of these enzyme deficiencies what the tissue specific roles of the pathway are compared to secondary systemic effects. To understand the cell or tissue specific contributions of fatty acid oxidation to systemic physiology, conditional knockouts in mice have been employed to determine the requirements of fatty acid oxidation in disparate cell types. This has produced a host of surprising results that sometimes run counter to the canonical view of this metabolic pathway. The rigor of conditional knockouts has also provided clarity over previous research utilizing cell lines in vitro or small molecule inhibitors with dubious specificity. Here we will summarize current research using mouse models of Carnitine Palmitoyltransferases to determine the tissue specific roles and requirements of long chain mitochondrial fatty acid β-oxidation.
Collapse
Affiliation(s)
- Danielle M Smith
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joseph Choi
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael J Wolfgang
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
Axsom J, TeSlaa T, Lee WD, Chu Q, Cowan A, Bornstein MR, Neinast MD, Bartman CR, Blair MC, Li K, Thorsheim C, Rabinowitz JD, Arany Z. Quantification of nutrient fluxes during acute exercise in mice. Cell Metab 2024; 36:2560-2579.e5. [PMID: 39413791 PMCID: PMC11620932 DOI: 10.1016/j.cmet.2024.09.010] [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: 07/13/2023] [Revised: 05/03/2024] [Accepted: 09/17/2024] [Indexed: 10/18/2024]
Abstract
Despite the known metabolic benefits of exercise, an integrated metabolic understanding of exercise is lacking. Here, we use in vivo steady-state isotope-labeled infusions to quantify fuel flux and oxidation during exercise in fasted, fed, and exhausted female mice, revealing several novel findings. Exercise strongly promoted glucose fluxes from liver glycogen, lactate, and glycerol, distinct from humans. Several organs spared glucose, a process that broke down in exhausted mice despite concomitant hypoglycemia. Proteolysis increased markedly, also divergent from humans. Fatty acid oxidation dominated during fasted exercise. Ketone production and oxidation rose rapidly, seemingly driven by a hepatic bottleneck caused by gluconeogenesis-induced cataplerotic stress. Altered fuel consumption was observed in organs not directly involved in muscle contraction, including the pancreas and brown fat. Several futile cycles surprisingly persisted during exercise, despite their energy cost. In sum, we provide a comprehensive, integrated, holistic, and quantitative accounting of metabolism during exercise in an intact organism.
Collapse
Affiliation(s)
- Jessie Axsom
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tara TeSlaa
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Won Dong Lee
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Qingwei Chu
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexis Cowan
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Marc R Bornstein
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael D Neinast
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Caroline R Bartman
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Megan C Blair
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristina Li
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chelsea Thorsheim
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Zoltan Arany
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Andreas E, Penn A, Okada T, St. John JC. Supplementation of Oocytes by Microinjection with Extra Copies of mtDNA Alters Metabolite Profiles and Interactions with Expressed Genes in a Tissue-Specific Manner. Biomolecules 2024; 14:1477. [PMID: 39595653 PMCID: PMC11591607 DOI: 10.3390/biom14111477] [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] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Mitochondrial DNA (mtDNA) supplementation can rescue poor oocyte quality and overcome embryonic arrest. Here, we investigated a series of sexually mature pigs generated through autologous and heterologous mtDNA supplementation. Brain, liver and heart tissues underwent metabolite profiling using gas chromatography-mass spectrometry and gene expression analysis through RNA-seq. They were then assessed for mRNA-metabolite interactions. The comparison between overall mtDNA supplemented and control pigs revealed that mtDNA supplementation reduced the lipids stearic acid and elaidic acid in heart tissue. However, heterologous mtDNA supplemented-derived pigs exhibited lower levels of abundance of metabolites when compared with autologous-derived pigs. In the brain, these included mannose, mannose 6-phosphate and fructose 6-phosphate. In the liver, maltose and cellobiose, and in the heart, glycine and glutamate were affected. mRNA-metabolite pathway analysis revealed a correlation between malate and CS, ACLY, IDH2 and PKLR in the liver and glutamate and PSAT1, PHGDH, CDO1 and ANPEP in the heart. Our outcomes demonstrate that mtDNA supplementation, especially heterologous supplementation, alters the metabolite and transcriptome profiles of brain, liver, and heart tissues. This is likely due to the extensive resetting of the balance between the nuclear and mitochondrial genomes in the preimplantation embryo, which induces a series of downstream effects.
Collapse
Affiliation(s)
| | | | | | - Justin C. St. John
- Experimental Mitochondrial Genetics Group, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide Health and Medical Sciences Building, Adelaide, SA 5000, Australia; (E.A.); (A.P.); (T.O.)
| |
Collapse
|
17
|
Nuyttens L, Vandewalle J, Libert C. Sepsis-induced changes in pyruvate metabolism: insights and potential therapeutic approaches. EMBO Mol Med 2024; 16:2678-2698. [PMID: 39468303 PMCID: PMC11554794 DOI: 10.1038/s44321-024-00155-6] [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: 06/10/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
Sepsis is a heterogeneous syndrome resulting from a dysregulated host response to infection. It is considered as a global major health priority. Sepsis is characterized by significant metabolic perturbations, leading to increased circulating metabolites such as lactate. In mammals, pyruvate is the primary substrate for lactate production. It plays a critical role in metabolism by linking glycolysis, where it is produced, with the mitochondrial oxidative phosphorylation pathway, where it is oxidized. Here, we provide an overview of all cytosolic and mitochondrial enzymes involved in pyruvate metabolism and how their activities are disrupted in sepsis. Based on the available data, we also discuss potential therapeutic strategies targeting these pyruvate-related enzymes leading to enhanced survival.
Collapse
Affiliation(s)
- Louise Nuyttens
- Center for Inflammation Research, Vlaams Instituut voor Biotechnologie (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jolien Vandewalle
- Center for Inflammation Research, Vlaams Instituut voor Biotechnologie (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Claude Libert
- Center for Inflammation Research, Vlaams Instituut voor Biotechnologie (VIB), Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
18
|
Yu HC, Tseng HYH, Huang HB, Lu MC. Circ-CAMTA1 regulated by Ca 2+ influx inhibited pyruvate carboxylase activity and modulate T cell function in patients with systemic lupus erythematosus. Arthritis Res Ther 2024; 26:185. [PMID: 39473004 PMCID: PMC11520813 DOI: 10.1186/s13075-024-03422-6] [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: 07/31/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
OBJECTIVES To investigate the roles of Ca2+ influx-regulated circular RNAs (circRNAs) in T cells from patients with systemic lupus erythematosus (SLE). METHODS The expression profile of circRNAs in Jurkat cells, co-cultured with and without ionomycin, was analyzed by next-generation sequencing and validated using real-time polymerase chain reaction. The identified Ca2+ influx-regulated circRNAs were further examined in T cells from 42 patients with SLE and 23 healthy controls. The biological function of specific circRNA was investigated using transfection and RNA pull-down assay. RESULTS After validation, we confirmed that the expression levels of circ-ERCC4, circ-NFATC2, circ-MYH10, circ-CAMTA1, circ-ASH1L, circ-SOCS7, and circ-ASAP1 were consistently increased in Jurkat cells following Ca2+ influx. The expression levels of circ-CAMTA1, circ-ASH1L, and circ-ASAP1 were significantly lower in T cells from patients with SLE, with even lower levels observed in those with higher disease activity. Interferon (IFN)-α was found to suppress the expression of circ-CAMTA1. Circ-CAMTA1 bound to pyruvate carboxylase and inhibited its biological activity. Overexpression of circ-CAMTA1, but not its linear form, significantly decreased extracellular glucose levels. Furthermore, increased expression of circ-CAMTA1, but not its linear form, decreased miR-181c-5p expression, resulting increased IL-2 secretion. CONCLUSION Three Ca2+ influx-regulated circ-RNAs-circ-CAMTA1, circ-ASH1L, and circ-ASAP1 -were significantly reduced in T cells from patients with SLE and associated with disease activity. IFN-α suppressed the expression of circ-CAMTA1, which interacted with pyruvate carboxylase, inhibited its activity, affected glucose metabolism, and increased IL-2 secretion. These findings suggest that circ-CAMTA1 regulated by Ca²⁺ influx modulated T cell function in patients with SLE.
Collapse
Affiliation(s)
- Hui-Chun Yu
- Division of Allergy, Immunology and Rheumatology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 2, Minsheng Road, Dalin, Chiayi, 62247, Taiwan
| | - Hsien-Yu Huang Tseng
- Division of Allergy, Immunology and Rheumatology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 2, Minsheng Road, Dalin, Chiayi, 62247, Taiwan
| | - Hsien-Bin Huang
- Department of Life Science, Institute of Molecular Biology, National Chung Cheng University, Minxiong, Chiayi, Taiwan
| | - Ming-Chi Lu
- Division of Allergy, Immunology and Rheumatology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 2, Minsheng Road, Dalin, Chiayi, 62247, Taiwan.
- School of Medicine, Tzu Chi University, Hualien City, Taiwan.
| |
Collapse
|
19
|
Li N, Li H, Chen Z, Feng J, Guo T, Guo H, Zhang X, Yan Y, He C, Zong D. Transcriptome and Metabolome Based Mechanisms Revealing the Accumulation and Transformation of Sugars and Fats in Pinus armandii Seed Kernels during the Harvesting Period. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21533-21547. [PMID: 39306861 DOI: 10.1021/acs.jafc.4c03100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Pinus armandii seed kernel is a nutrient-rich and widely consumed nut whose yield and quality are affected by, among other things, harvesting time and climatic conditions, which reduce economic benefits. To investigate the optimal harvesting period of P. armandii seed kernels, this study determined the nutrient composition and seed kernel morphology and analyzed the gene expression and metabolic differences of P. armandii seed kernels during the harvesting period by transcriptomics and metabolomics. The results revealed that during the maturation of P. armandii seed kernels, there was a significant increase in the width, thickness, and weight of the seed kernels, as well as a significant accumulation of sucrose, soluble sugars, proteins, starch, flavonoids, and polyphenols and a significant decrease in lipid content. In addition, transcriptomic and metabolomic analyses of P. armandii seed kernels during the harvesting period screened and identified 103 differential metabolites (DEMs) and 8899 differential genes (DEGs). Analysis of these DEMs and DEGs revealed that P. armandii seed kernel harvesting exhibited gene-metabolite differences in sugar- and lipid-related pathways. Among them, starch and sucrose metabolism, glycolysis, and gluconeogenesis were associated with the synthesis and catabolism of sugars, whereas fatty acid degradation, glyoxylate and dicarboxylic acid metabolism, and glycerophospholipid metabolism were associated with the synthesis and catabolism of lipids. Therefore, the present study hypothesized that these differences in genes and metabolites exhibited during the harvesting period of P. armandii seed kernels might be related to the accumulation and transformation of sugars and lipids. This study may provide a theoretical basis for determining the optimal harvesting time of P. armandii seed kernels, changes in the molecular mechanisms of nutrient accumulation, and quality directed breeding.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory for Forest Genetic and Tree Improvement &Propagation in Universities of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Key Laboratory of State Forestry Administration on Biodiversity Conservation in Southwest China, Southwest Forestry University, Kunming 650224, China
| | - Hailin Li
- Key Laboratory for Forest Genetic and Tree Improvement &Propagation in Universities of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Key Laboratory of State Forestry Administration on Biodiversity Conservation in Southwest China, Southwest Forestry University, Kunming 650224, China
- Key Laboratory for Forest Resources Conservation and Use in the Southwest Mountains of China, Ministry of Education, Southwest Forestry University, Kunming 650224, China
| | - Zhihua Chen
- Key Laboratory for Forest Genetic and Tree Improvement &Propagation in Universities of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Key Laboratory of State Forestry Administration on Biodiversity Conservation in Southwest China, Southwest Forestry University, Kunming 650224, China
| | - Jiayu Feng
- Key Laboratory for Forest Genetic and Tree Improvement &Propagation in Universities of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Key Laboratory of State Forestry Administration on Biodiversity Conservation in Southwest China, Southwest Forestry University, Kunming 650224, China
| | - Tiansu Guo
- Key Laboratory for Forest Genetic and Tree Improvement &Propagation in Universities of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Key Laboratory of State Forestry Administration on Biodiversity Conservation in Southwest China, Southwest Forestry University, Kunming 650224, China
| | - Haiyang Guo
- Key Laboratory for Forest Genetic and Tree Improvement &Propagation in Universities of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Key Laboratory of State Forestry Administration on Biodiversity Conservation in Southwest China, Southwest Forestry University, Kunming 650224, China
| | - Xiaolin Zhang
- Key Laboratory for Forest Genetic and Tree Improvement &Propagation in Universities of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Key Laboratory of State Forestry Administration on Biodiversity Conservation in Southwest China, Southwest Forestry University, Kunming 650224, China
| | - Yi Yan
- Kunming Forestry Scientific Research Institute, Kunming 650221, China
| | - Chengzhong He
- Key Laboratory for Forest Genetic and Tree Improvement &Propagation in Universities of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Key Laboratory of State Forestry Administration on Biodiversity Conservation in Southwest China, Southwest Forestry University, Kunming 650224, China
- Key Laboratory for Forest Resources Conservation and Use in the Southwest Mountains of China, Ministry of Education, Southwest Forestry University, Kunming 650224, China
| | - Dan Zong
- Key Laboratory for Forest Genetic and Tree Improvement &Propagation in Universities of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Key Laboratory of State Forestry Administration on Biodiversity Conservation in Southwest China, Southwest Forestry University, Kunming 650224, China
- Key Laboratory for Forest Resources Conservation and Use in the Southwest Mountains of China, Ministry of Education, Southwest Forestry University, Kunming 650224, China
| |
Collapse
|
20
|
Renoult O, Laurent--Blond M, Awada H, Oliver L, Joalland N, Croyal M, Paris F, Gratas C, Pecqueur C. Metabolic profiling of glioblastoma stem cells reveals pyruvate carboxylase as a critical survival factor and potential therapeutic target. Neuro Oncol 2024; 26:1572-1586. [PMID: 38869884 PMCID: PMC11376449 DOI: 10.1093/neuonc/noae106] [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: 09/19/2023] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive tumor with unmet therapeutic needs, which can be explained by extensive intra-tumoral heterogeneity and plasticity. In this study, we aimed to investigate the specific metabolic features of Glioblastoma stem cells (GSC), a rare tumor subpopulation involved in tumor growth and therapy resistance. METHODS We conducted comprehensive analyses of primary patient-derived GBM cultures and GSC-enriched cultures of human GBM cell lines using state-of-the-art molecular, metabolic, and phenotypic studies. RESULTS We showed that GSC-enriched cultures display distinct glycolytic profiles compared with differentiated tumor cells. Further analysis revealed that GSC relies on pyruvate carboxylase (PC) activity for survival and self-renewal capacity. Interestingly, inhibition of PC led to GSC death, particularly when the glutamine pool was low, and increased differentiation. Finally, while GSC displayed resistance to the chemotherapy drug etoposide, genetic or pharmacological inhibition of PC restored etoposide sensitivity in GSC, both in vitro and in orthotopic murine models. CONCLUSIONS Our findings demonstrate the critical role of PC in GSC metabolism, survival, and escape to etoposide. They also highlight PC as a therapeutic target to overcome therapy resistance in GBM.
Collapse
Affiliation(s)
- Ophélie Renoult
- Nantes Université, Inserm 1307, CNRS 6075, Université d’Angers, Nantes, France
| | | | - Hala Awada
- Faculty of Sciences, Lebanese University, Beirut, Lebanon
- Nantes Université, Inserm 1307, CNRS 6075, Université d’Angers, Nantes, France
| | - Lisa Oliver
- Centre Hospitalier Universitaire de Nantes, Nantes, France
- Nantes Université, Inserm 1307, CNRS 6075, Université d’Angers, Nantes, France
| | - Noémie Joalland
- Nantes Université, Inserm 1307, CNRS 6075, Université d’Angers, Nantes, France
| | - Mikaël Croyal
- Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
- Université de Nantes, CNRS, INSERM, l’institut du thorax, Nantes, France
| | - François Paris
- Institut de Cancérologie de l’Ouest, Saint-Herblain, France
- Nantes Université, Inserm 1307, CNRS 6075, Université d’Angers, Nantes, France
| | - Catherine Gratas
- Centre Hospitalier Universitaire de Nantes, Nantes, France
- Nantes Université, Inserm 1307, CNRS 6075, Université d’Angers, Nantes, France
| | - Claire Pecqueur
- Nantes Université, Inserm 1307, CNRS 6075, Université d’Angers, Nantes, France
| |
Collapse
|
21
|
Taha M, Assali EA, Ben-Kasus Nissim T, Stutzmann GE, Shirihai OS, Hershfinkel M, Sekler I. NCLX controls hepatic mitochondrial Ca 2+ extrusion and couples hormone-mediated mitochondrial Ca 2+ oscillations with gluconeogenesis. Mol Metab 2024; 87:101982. [PMID: 38960129 PMCID: PMC11325370 DOI: 10.1016/j.molmet.2024.101982] [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: 03/27/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
OBJECTIVE Hepatic Ca2+ signaling has been identified as a crucial key factor in driving gluconeogenesis. The involvement of mitochondria in hormone-induced Ca2+ signaling and their contribution to metabolic activity remain, however, poorly understood. Moreover, the molecular mechanism governing the mitochondrial Ca2+ efflux signaling remains unresolved. This study investigates the role of the Na+/Ca2+ exchanger, NCLX, in modulating hepatic mitochondrial Ca2+ efflux, and examines its physiological significance in hormonal hepatic Ca2+ signaling, gluconeogenesis, and mitochondrial bioenergetics. METHODS Primary mouse hepatocytes from both an AAV-mediated conditional hepatic-specific and a total mitochondrial Na+/Ca2+ exchanger, NCLX, knockout (KO) mouse models were employed for fluorescent monitoring of purinergic and glucagon/vasopressin-dependent mitochondrial and cytosolic hepatic Ca2+ responses in cultured hepatocytes. Isolated liver mitochondria and permeabilized primary hepatocytes were used to analyze the ion-dependence of Ca2+ efflux. Utilizing the conditional hepatic-specific NCLX KO model, the rate of gluconeogenesis was assessed by first monitoring glucose levels in fasted mice, and subsequently subjecting the mice to a pyruvate tolerance test while monitoring their blood glucose. Additionally, cultured primary hepatocytes from both genotypes were assessed in vitro for glucagon-dependent glucose production and cellular bioenergetics through glucose oxidase assay and Seahorse respirometry, respectively. RESULTS Analysis of Ca2+ responses in isolated liver mitochondria and cultured primary hepatocytes from NCLX KO versus WT mice showed that NCLX serves as the principal mechanism for mitochondrial calcium extrusion in hepatocytes. We then determined the role of NCLX in glucagon and vasopressin-induced Ca2+ oscillations. Consistent with previous studies, glucagon and vasopressin triggered Ca2+ oscillations in WT hepatocytes, however, the deletion of NCLX resulted in selective elimination of mitochondrial, but not cytosolic, Ca2+ oscillations, underscoring NCLX's pivotal role in mitochondrial Ca2+ regulation. Subsequent in vivo investigation for hepatic NCLX role in gluconeogenesis revealed that, as opposed to WT mice which maintained normoglycemic blood glucose levels when fasted, conditional hepatic-specific NCLX KO mice exhibited a faster drop in glucose levels, becoming hypoglycemic. Furthermore, KO mice showed deficient conversion of pyruvate to glucose when challenged under fasting conditions. Concurrent in vitro assessments showed impaired glucagon-dependent glucose production and compromised bioenergetics in KO hepatocytes, thereby underscoring NCLX's significant contribution to hepatic glucose metabolism. CONCLUSIONS The study findings demonstrate that NCLX acts as the primary Ca2+ efflux mechanism in hepatocytes. NCLX is indispensable for regulating hormone-induced mitochondrial Ca2+ oscillations, mitochondrial metabolism, and sustenance of hepatic gluconeogenesis.
Collapse
Affiliation(s)
- Mahmoud Taha
- Department of Physiology and Cell Biology, Ben Gurion University, Beer-Sheva 8410501, Israel
| | - Essam A Assali
- Department of Physiology and Cell Biology, Ben Gurion University, Beer-Sheva 8410501, Israel; Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| | - Tsipi Ben-Kasus Nissim
- Department of Physiology and Cell Biology, Ben Gurion University, Beer-Sheva 8410501, Israel
| | - Grace E Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science. North Chicago, IL 60064, USA
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology, Ben Gurion University, Beer-Sheva 8410501, Israel
| | - Israel Sekler
- Department of Physiology and Cell Biology, Ben Gurion University, Beer-Sheva 8410501, Israel.
| |
Collapse
|
22
|
Marumo T, Yoshida N, Inoue N, Yamanouchi M, Ubara Y, Urakami S, Fujii T, Takazawa Y, Ohashi K, Kawarazaki W, Nishimoto M, Ayuzawa N, Hirohama D, Nagae G, Fujimoto M, Arai E, Kanai Y, Hoshino J, Fujita T. Aberrant proximal tubule DNA methylation underlies phenotypic changes related to kidney dysfunction in patients with diabetes. Am J Physiol Renal Physiol 2024; 327:F397-F411. [PMID: 38961842 DOI: 10.1152/ajprenal.00124.2024] [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: 04/22/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
Epigenetic mechanisms are considered to contribute to diabetic nephropathy by maintaining memory of poor glycemic control during the early stages of diabetes. However, DNA methylation changes in the human kidney are poorly characterized, because of the lack of cell type-specific analysis. We examined DNA methylation in proximal tubules (PTs) purified from patients with diabetic nephropathy and identified differentially methylated CpG sites, given the critical role of proximal tubules in the kidney injury. Hypermethylation was observed at CpG sites annotated to genes responsible for proximal tubule functions, including gluconeogenesis, nicotinamide adenine dinucleotide synthesis, transporters of glucose, water, phosphate, and drugs, in diabetic kidneys, whereas genes involved in oxidative stress and the cytoskeleton exhibited demethylation. Methylation levels of CpG sites annotated to ACTN1, BCAR1, MYH9, UBE4B, AFMID, TRAF2, TXNIP, FOXO3, and HNF4A were correlated with the estimated glomerular filtration rate, whereas methylation of the CpG site in RUNX1 was associated with interstitial fibrosis and tubular atrophy. Hypermethylation of G6PC and HNF4A was accompanied by decreased expression in diabetic kidneys. Proximal tubule-specific hypomethylation of metabolic genes related to HNF4A observed in control kidneys was compromised in diabetic kidneys, suggesting a role for aberrant DNA methylation in the dedifferentiation process. Multiple genes with aberrant DNA methylation in diabetes overlapped genes with altered expressions in maladaptive proximal tubule cells, including transcription factors PPARA and RREB1. In conclusion, DNA methylation derangement in the proximal tubules of patients with diabetes may drive phenotypic changes, characterized by inflammatory and fibrotic features, along with impaired function in metabolism and transport.NEW & NOTEWORTHY Cell type-specific DNA methylation patterns in the human kidney are not known. We examined DNA methylation in proximal tubules of patients with diabetic nephropathy and revealed that oxidative stress, cytoskeleton, and metabolism genes were aberrantly methylated. The results indicate that aberrant DNA methylation in proximal tubules underlies kidney dysfunction in diabetic nephropathy. Aberrant methylation could be a target for reversing memory of poor glycemic control.
Collapse
Affiliation(s)
- Takeshi Marumo
- Department of Pharmacology, School of Medicine, International University of Health and Welfare, Chiba, Japan
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Naoto Yoshida
- Department of Pharmacology, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Noriko Inoue
- Nephrology Center, Toranomon Hospital, Tokyo, Japan
| | | | | | | | - Takeshi Fujii
- Department of Pathology, Toranomon Hospital, Tokyo, Japan
| | | | - Kenichi Ohashi
- Department of Human Pathology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Wakako Kawarazaki
- Department of Pharmacology, School of Medicine, International University of Health and Welfare, Chiba, Japan
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Mitsuhiro Nishimoto
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Nobuhiro Ayuzawa
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Daigoro Hirohama
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Genta Nagae
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Mao Fujimoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Eri Arai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Junichi Hoshino
- Nephrology Center, Toranomon Hospital, Tokyo, Japan
- Deparment of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshiro Fujita
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Phomvisith O, Muroya S, Otomaru K, Oshima K, Oshima I, Nishino D, Haginouchi T, Gotoh T. Maternal Undernutrition Affects Fetal Thymus DNA Methylation, Gene Expression, and, Thereby, Metabolism and Immunopoiesis in Wagyu (Japanese Black) Cattle. Int J Mol Sci 2024; 25:9242. [PMID: 39273192 PMCID: PMC11395129 DOI: 10.3390/ijms25179242] [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: 07/30/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
We aimed to determine the effects of maternal nutrient restriction (MNR) on the DNA methylation and gene expression patterns associated with metabolism and immunopoiesis in the thymuses of fetal Wagyu cattle. Pregnant cows were allocated to two groups: a low-nutrition (LN; 60% nutritional requirement; n = 5) and a high-nutrition (HN; 120% nutritional requirement, n = 6) group, until 8.5 months of gestation. Whole-genome bisulfite sequencing (WGBS) and RNA sequencing were used to analyze DNA methylation and gene expression, while capillary electrophoresis-Fourier transform mass spectrometry assessed the metabolome. WGBS identified 4566 hypomethylated and 4303 hypermethylated genes in the LN group, with the intergenic regions most frequently being methylated. Pathway analysis linked hypoDMGs to Ras signaling, while hyperDMGs were associated with Hippo signaling. RNA sequencing found 94 differentially expressed genes (66 upregulated, 28 downregulated) in the LN group. The upregulated genes were tied to metabolic pathways and oxidative phosphorylation; the downregulated genes were linked to natural killer cell cytotoxicity. Key overlapping genes (GRIA1, CACNA1D, SCL25A4) were involved in cAMP signaling. The metabolomic analysis indicated an altered amino acid metabolism in the MNR fetuses. These findings suggest that MNR affects DNA methylation, gene expression, and the amino acid metabolism, impacting immune system regulation during fetal thymus development in Wagyu cattle.
Collapse
Affiliation(s)
- Ouanh Phomvisith
- Field Science Center for Northern Biosphere, Hokkaido University, N11W10, Kita, Sapporo 060-0811, Hokkaido, Japan
| | - Susumu Muroya
- Department of Animal Science, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Kagoshima, Japan
| | - Konosuke Otomaru
- Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Kagoshima, Japan
| | - Kazunaga Oshima
- Division of Year-Round Grazing Research, NARO Western Region Agricultural Research Center, 60 Yoshinaga, Ohda 694-0013, Shimane, Japan
| | - Ichiro Oshima
- Department of Animal Science, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Kagoshima, Japan
| | - Daichi Nishino
- Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 744 Motooka Nishi-ku, Fukuoka 819-0395, Fukuoka, Japan
| | - Taketo Haginouchi
- Field Science Center for Northern Biosphere, Hokkaido University, N11W10, Kita, Sapporo 060-0811, Hokkaido, Japan
| | - Takafumi Gotoh
- Field Science Center for Northern Biosphere, Hokkaido University, N11W10, Kita, Sapporo 060-0811, Hokkaido, Japan
| |
Collapse
|
24
|
Wang Z, Lan H, Wang Y, Zheng Q, Li C, Wang K, Xiong T, Wu Q, Dong N. Pyruvate Carboxylase Attenuates Myocardial Ischemia-Reperfusion Injury in Heart Transplantation via Wnt/β-Catenin-Mediated Glutamine Metabolism. Biomedicines 2024; 12:1826. [PMID: 39200290 PMCID: PMC11351651 DOI: 10.3390/biomedicines12081826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
The ischemia-reperfusion process of a donor heart during heart transplantation leads to severe mitochondrial dysfunction, which may be the main cause of donor heart dysfunction after heart transplantation. Pyruvate carboxylase (PC), an enzyme found in mitochondria, is said to play a role in the control of oxidative stress and the function of mitochondria. This research examined the function of PC and discovered the signaling pathways controlled by PC in myocardial IRI. We induced IRI using a murine heterotopic heart transplantation model in vivo and a hypoxia-reoxygenation cell model in vitro and evaluated inflammatory responses, oxidative stress levels, mitochondrial function, and cardiomyocyte apoptosis. In both in vivo and in vitro settings, we observed a significant decrease in PC expression during myocardial IRI. PC knockdown aggravated IRI by increasing MDA content, LDH activity, TUNEL-positive cells, serum cTnI level, Bax protein expression, and the level of inflammatory cytokines and decreasing SOD activity, GPX activity, and Bcl-2 protein expression. PC overexpression yielded the opposite findings. Additional research indicated that reducing PC levels could block the Wnt/β-catenin pathway and glutamine metabolism by hindering the movement of β-catenin to the nucleus and reducing the activity of complex I and complex II, as well as ATP levels, while elevating the ratios of NADP+/NADPH and GSSG/GSH. Overall, the findings indicated that PC therapy can shield the heart from IRI during heart transplantation by regulating glutamine metabolism through the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Zihao Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China; (Z.W.)
| | - Hongwen Lan
- Department of Thoracic Surgery, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China; (Z.W.)
| | - Qiang Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China; (Z.W.)
| | - Chenghao Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China; (Z.W.)
| | - Kan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China; (Z.W.)
| | - Tixiusi Xiong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China; (Z.W.)
| | - Qingping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China; (Z.W.)
| |
Collapse
|
25
|
Jacquemin C, El Orch W, Diaz O, Lalande A, Aublin-Gex A, Jacolin F, Toesca J, Si-Tahar M, Mathieu C, Lotteau V, Perrin-Cocon L, Vidalain PO. Pharmacological induction of the hypoxia response pathway in Huh7 hepatoma cells limits proliferation but increases resilience under metabolic stress. Cell Mol Life Sci 2024; 81:320. [PMID: 39078527 PMCID: PMC11335246 DOI: 10.1007/s00018-024-05361-6] [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: 04/02/2024] [Revised: 06/25/2024] [Accepted: 07/12/2024] [Indexed: 07/31/2024]
Abstract
The hypoxia response pathway enables adaptation to oxygen deprivation. It is mediated by hypoxia-inducible factors (HIF), which promote metabolic reprogramming, erythropoiesis, angiogenesis and tissue remodeling. This led to the successful development of HIF-inducing drugs for treating anemia and some of these molecules are now in clinic. However, elevated levels of HIFs are frequently associated with tumor growth, poor prognosis, and drug resistance in various cancers, including hepatocellular carcinoma (HCC). Consequently, there are concerns regarding the recommendation of HIF-inducing drugs in certain clinical situations. Here, we analyzed the effects of two HIF-inducing drugs, Molidustat and Roxadustat, in the well-characterized HCC cell line Huh7. These drugs increased HIF-1α and HIF-2α protein levels which both participate in inducing hypoxia response genes such as BNIP3, SERPINE1, LDHA or EPO. Combined transcriptomics, proteomics and metabolomics showed that Molidustat increased the expression of glycolytic enzymes, while the mitochondrial network was fragmented and cellular respiration decreased. This metabolic remodeling was associated with a reduced proliferation and a lower demand for pyrimidine supply, but an increased ability of cells to convert pyruvate to lactate. This was accompanied by a higher resistance to the inhibition of mitochondrial respiration by antimycin A, a phenotype confirmed in Roxadustat-treated Huh7 cells and Molidustat-treated hepatoblastoma cells (Huh6 and HepG2). Overall, this study shows that HIF-inducing drugs increase the metabolic resilience of liver cancer cells to metabolic stressors, arguing for careful monitoring of patients treated with HIF-inducing drugs, especially when they are at risk of liver cancer.
Collapse
Affiliation(s)
- Clémence Jacquemin
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Walid El Orch
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Olivier Diaz
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Alexandre Lalande
- CIRI, Centre International de Recherche en Infectiologie, Team NeuroInvasion, Tropism and Viral Encephalitis, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Anne Aublin-Gex
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Florentine Jacolin
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Johan Toesca
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Mustapha Si-Tahar
- Centre d'Etude des Pathologies Respiratoires (CEPR), Faculty of Medecine, Inserm, U1100, 37000, Tours, France
| | - Cyrille Mathieu
- CIRI, Centre International de Recherche en Infectiologie, Team NeuroInvasion, Tropism and Viral Encephalitis, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Vincent Lotteau
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
- Laboratoire P4 INSERM-Jean Mérieux, Lyon, France
| | - Laure Perrin-Cocon
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France.
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France.
| |
Collapse
|
26
|
Elnwasany A, Ewida HA, Menendez-Montes I, Mizerska M, Fu X, Kim CW, Horton JD, Burgess SC, Rothermel BA, Szweda PA, Szweda LI. Reciprocal regulation of cardiac β-oxidation and pyruvate dehydrogenase by insulin. J Biol Chem 2024; 300:107412. [PMID: 38796064 PMCID: PMC11231754 DOI: 10.1016/j.jbc.2024.107412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
The heart alters the rate and relative oxidation of fatty acids and glucose based on availability and energetic demand. Insulin plays a crucial role in this process diminishing fatty acid and increasing glucose oxidation when glucose availability increases. Loss of insulin sensitivity and metabolic flexibility can result in cardiovascular disease. It is therefore important to identify mechanisms by which insulin regulates substrate utilization in the heart. Mitochondrial pyruvate dehydrogenase (PDH) is the key regulatory site for the oxidation of glucose for ATP production. Nevertheless, the impact of insulin on PDH activity has not been fully delineated, particularly in the heart. We sought in vivo evidence that insulin stimulates cardiac PDH and that this process is driven by the inhibition of fatty acid oxidation. Mice injected with insulin exhibited dephosphorylation and activation of cardiac PDH. This was accompanied by an increase in the content of malonyl-CoA, an inhibitor of carnitine palmitoyltransferase 1 (CPT1), and, thus, mitochondrial import of fatty acids. Administration of the CPT1 inhibitor oxfenicine was sufficient to activate PDH. Malonyl-CoA is produced by acetyl-CoA carboxylase (ACC). Pharmacologic inhibition or knockout of cardiac ACC diminished insulin-dependent production of malonyl-CoA and activation of PDH. Finally, circulating insulin and cardiac glucose utilization exhibit daily rhythms reflective of nutritional status. We demonstrate that time-of-day-dependent changes in PDH activity are mediated, in part, by ACC-dependent production of malonyl-CoA. Thus, by inhibiting fatty acid oxidation, insulin reciprocally activates PDH. These studies identify potential molecular targets to promote cardiac glucose oxidation and treat heart disease.
Collapse
Affiliation(s)
- Abdallah Elnwasany
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Heba A Ewida
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, USA; Faculty of Pharmacy, Future University in Egypt (FUE), Cairo, Egypt
| | - Ivan Menendez-Montes
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Monika Mizerska
- Department of Pharmacology, Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xiaorong Fu
- Department of Pharmacology, Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chai-Wan Kim
- Departments of Internal Medicine and Molecular Genetics, Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jay D Horton
- Departments of Internal Medicine and Molecular Genetics, Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Shawn C Burgess
- Department of Pharmacology, Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Beverly A Rothermel
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Pamela A Szweda
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Luke I Szweda
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
27
|
Timpani CA, Debrincat D, Kourakis S, Boyer R, Formosa LE, Steele JR, Zhang H, Schittenhelm RB, Russell AP, Rybalka E, Lindsay A. Loss of endogenous estrogen alters mitochondrial metabolism and muscle clock-related protein Rbm20 in female mdx mice. FASEB J 2024; 38:e23718. [PMID: 38847487 DOI: 10.1096/fj.202400329r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/08/2024] [Accepted: 05/19/2024] [Indexed: 11/01/2024]
Abstract
Female carriers of a Duchenne muscular dystrophy (DMD) gene mutation manifest exercise intolerance and metabolic anomalies that may be exacerbated following menopause due to the loss of estrogen, a known regulator of skeletal muscle function and metabolism. Here, we studied the impact of estrogen depletion (via ovariectomy) on exercise tolerance and muscle mitochondrial metabolism in female mdx mice and the potential of estrogen replacement therapy (using estradiol) to protect against functional and metabolic perturbations. We also investigated the effect of estrogen depletion, and replacement, on the skeletal muscle proteome through an untargeted proteomic approach with TMT-labelling. Our study confirms that loss of estrogen in female mdx mice reduces exercise capacity, tricarboxylic acid cycle intermediates, and citrate synthase activity but that these deficits are offset through estrogen replacement therapy. Furthermore, ovariectomy downregulated protein expression of RNA-binding motif factor 20 (Rbm20), a critical regulator of sarcomeric and muscle homeostasis gene splicing, which impacted pathways involving ribosomal and mitochondrial translation. Estrogen replacement modulated Rbm20 protein expression and promoted metabolic processes and the upregulation of proteins involved in mitochondrial dynamics and metabolism. Our data suggest that estrogen mitigates dystrophinopathic features in female mdx mice and that estrogen replacement may be a potential therapy for post-menopausal DMD carriers.
Collapse
Affiliation(s)
- Cara A Timpani
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
| | - Didier Debrincat
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Stephanie Kourakis
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Rebecca Boyer
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Luke E Formosa
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Joel R Steele
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Haijian Zhang
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Aaron P Russell
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Emma Rybalka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
- Division of Neuropaediatrics and Developmental Medicine, University Children's Hospital of Basel (UKBB), Basel, Switzerland
| | - Angus Lindsay
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
- Department of Medicine, University of Otago, Christchurch, New Zealand
| |
Collapse
|
28
|
Bednarski TK, Rahim M, Hasenour CM, Banerjee DR, Trenary IA, Wasserman DH, Young JD. Pharmacological SERCA activation limits diet-induced steatohepatitis and restores liver metabolic function in mice. J Lipid Res 2024; 65:100558. [PMID: 38729350 PMCID: PMC11179628 DOI: 10.1016/j.jlr.2024.100558] [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: 01/25/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/12/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease is the most common form of liver disease and poses significant health risks to patients who progress to metabolic dysfunction-associated steatohepatitis. Fatty acid overload alters endoplasmic reticulum (ER) calcium stores and induces mitochondrial oxidative stress in hepatocytes, leading to hepatocellular inflammation and apoptosis. Obese mice have impaired liver sarco/ER Ca2+-ATPase (SERCA) function, which normally maintains intracellular calcium homeostasis by transporting Ca2+ ions from the cytoplasm to the ER. We hypothesized that restoration of SERCA activity would improve diet-induced steatohepatitis in mice by limiting ER stress and mitochondrial dysfunction. WT and melanocortin-4 receptor KO (Mc4r-/-) mice were placed on either chow or Western diet (WD) for 8 weeks. Half of the WD-fed mice were administered CDN1163 to activate SERCA, which reduced liver fibrosis and inflammation. SERCA activation also restored glucose tolerance and insulin sensitivity, improved histological markers of metabolic dysfunction-associated steatohepatitis, increased expression of antioxidant enzymes, and decreased expression of oxidative stress and ER stress genes. CDN1163 decreased hepatic citric acid cycle flux and liver pyruvate cycling, enhanced expression of mitochondrial respiratory genes, and shifted hepatocellular [NADH]/[NAD+] and [NADPH]/[NADP+] ratios to a less oxidized state, which was associated with elevated PUFA content of liver lipids. In sum, the data demonstrate that pharmacological SERCA activation limits metabolic dysfunction-associated steatotic liver disease progression and prevents metabolic dysfunction induced by WD feeding in mice.
Collapse
Affiliation(s)
- Tomasz K Bednarski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Clinton M Hasenour
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Deveena R Banerjee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Irina A Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
29
|
Li X, Pu Q, Xu Y, Yang H, Wu Y, Wang W, Li Y. The masking phenomenon of microplastics additives on oxidative stress responses in freshwater food chains. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:172156. [PMID: 38588742 DOI: 10.1016/j.scitotenv.2024.172156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/31/2024] [Accepted: 03/31/2024] [Indexed: 04/10/2024]
Abstract
The variability and intrinsic mechanisms of oxidative stress induced by microplastics at different trophic levels in freshwater food chains are not well understood. To comprehensively assess the oxidative stress induced by polystyrene microplastics (PS-MPs) in freshwater food chains, the present study first quantified the oxidative stress induced by PS-MPs in organisms at different trophic levels using factorial experimental design and molecular dynamics methods. Then focuses on analyzing the variability of these responses across different trophic levels using mathematical statistical analysis. Notably, higher trophic level organisms exhibit diminished responses under PS-MPs exposure. Furthermore, the coexistence of multiple additives was found to mask these responses, with antioxidant plastic additives significantly influencing oxidative stress responses. Mechanism analysis using computational chemistry simulation determines that protein structure and amino acid characteristics are key factors driving PS-MPs induced oxidative stress variation in freshwater organisms at different nutrient levels. Increased hydrophobic additives induce protein helicalization and amino acid residue aggregation. This study systematically reveals the variability of biological oxidative stress response under different nutrient levels, emphasizing the pivotal role of chemical additives. Overall, this study offers crucial insights into PS-MPs' impact on oxidative stress responses in freshwater ecosystems, informing future environmental risk assessment.
Collapse
Affiliation(s)
- Xinao Li
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China; MOE Key Laboratory of Resources and Environmental System Optimization, North China Electric Power University, Beijing 102206, China
| | - Qikun Pu
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China; MOE Key Laboratory of Resources and Environmental System Optimization, North China Electric Power University, Beijing 102206, China
| | - Yingjie Xu
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China; MOE Key Laboratory of Resources and Environmental System Optimization, North China Electric Power University, Beijing 102206, China
| | - Hao Yang
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China; MOE Key Laboratory of Resources and Environmental System Optimization, North China Electric Power University, Beijing 102206, China
| | - Yang Wu
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China; MOE Key Laboratory of Resources and Environmental System Optimization, North China Electric Power University, Beijing 102206, China
| | - Wenwen Wang
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China; MOE Key Laboratory of Resources and Environmental System Optimization, North China Electric Power University, Beijing 102206, China.
| | - Yu Li
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China; MOE Key Laboratory of Resources and Environmental System Optimization, North China Electric Power University, Beijing 102206, China.
| |
Collapse
|
30
|
Deja S, Fletcher JA, Kim CW, Kucejova B, Fu X, Mizerska M, Villegas M, Pudelko-Malik N, Browder N, Inigo-Vollmer M, Menezes CJ, Mishra P, Berglund ED, Browning JD, Thyfault JP, Young JD, Horton JD, Burgess SC. Hepatic malonyl-CoA synthesis restrains gluconeogenesis by suppressing fat oxidation, pyruvate carboxylation, and amino acid availability. Cell Metab 2024; 36:1088-1104.e12. [PMID: 38447582 PMCID: PMC11081827 DOI: 10.1016/j.cmet.2024.02.004] [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: 02/01/2023] [Revised: 12/10/2023] [Accepted: 02/09/2024] [Indexed: 03/08/2024]
Abstract
Acetyl-CoA carboxylase (ACC) promotes prandial liver metabolism by producing malonyl-CoA, a substrate for de novo lipogenesis and an inhibitor of CPT-1-mediated fat oxidation. We report that inhibition of ACC also produces unexpected secondary effects on metabolism. Liver-specific double ACC1/2 knockout (LDKO) or pharmacologic inhibition of ACC increased anaplerosis, tricarboxylic acid (TCA) cycle intermediates, and gluconeogenesis by activating hepatic CPT-1 and pyruvate carboxylase flux in the fed state. Fasting should have marginalized the role of ACC, but LDKO mice maintained elevated TCA cycle intermediates and preserved glycemia during fasting. These effects were accompanied by a compensatory induction of proteolysis and increased amino acid supply for gluconeogenesis, which was offset by increased protein synthesis during feeding. Such adaptations may be related to Nrf2 activity, which was induced by ACC inhibition and correlated with fasting amino acids. The findings reveal unexpected roles for malonyl-CoA synthesis in liver and provide insight into the broader effects of pharmacologic ACC inhibition.
Collapse
Affiliation(s)
- Stanislaw Deja
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Justin A Fletcher
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Chai-Wan Kim
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Blanka Kucejova
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Xiaorong Fu
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Monika Mizerska
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Morgan Villegas
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Natalia Pudelko-Malik
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Nicholas Browder
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Melissa Inigo-Vollmer
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Cameron J Menezes
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Prashant Mishra
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Eric D Berglund
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Jeffrey D Browning
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - John P Thyfault
- Departments of Cell Biology and Physiology, Internal Medicine and KU Diabetes Institute, Kansas Medical Center, Kansas City, KS, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| | - Jay D Horton
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA.
| | - Shawn C Burgess
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA.
| |
Collapse
|
31
|
Scoditti E, Sabatini S, Carli F, Gastaldelli A. Hepatic glucose metabolism in the steatotic liver. Nat Rev Gastroenterol Hepatol 2024; 21:319-334. [PMID: 38308003 DOI: 10.1038/s41575-023-00888-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 02/04/2024]
Abstract
The liver is central in regulating glucose homeostasis, being the major contributor to endogenous glucose production and the greatest reserve of glucose as glycogen. It is both a target and regulator of the action of glucoregulatory hormones. Hepatic metabolic functions are altered in and contribute to the highly prevalent steatotic liver disease (SLD), including metabolic dysfunction-associated SLD (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH). In this Review, we describe the dysregulation of hepatic glucose metabolism in MASLD and MASH and associated metabolic comorbidities, and how advances in techniques and models for the assessment of hepatic glucose fluxes in vivo have led to the identification of the mechanisms related to the alterations in glucose metabolism in MASLD and comorbidities. These fluxes can ultimately increase hepatic glucose production concomitantly with fat accumulation and alterations in the secretion and action of glucoregulatory hormones. No pharmacological treatment has yet been approved for MASLD or MASH, but some antihyperglycaemic drugs approved for treating type 2 diabetes have shown positive effects on hepatic glucose metabolism and hepatosteatosis. A deep understanding of how MASLD affects glucose metabolic fluxes and glucoregulatory hormones might assist in the early identification of at-risk individuals and the use or development of targeted therapies.
Collapse
Affiliation(s)
- Egeria Scoditti
- Institute of Clinical Physiology, National Research Council, Lecce, Italy
| | - Silvia Sabatini
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Fabrizia Carli
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council, Pisa, Italy.
| |
Collapse
|
32
|
Brown LD, Rozance PJ, Wang D, Eroglu EC, Wilkening RB, Solmonson A, Wesolowski SR. Increased hepatic glucose production with lower oxidative metabolism in the growth-restricted fetus. JCI Insight 2024; 9:e176497. [PMID: 38687612 PMCID: PMC11141920 DOI: 10.1172/jci.insight.176497] [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: 10/10/2023] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
Fetal growth restriction (FGR) is accompanied by early activation of hepatic glucose production (HGP), a hallmark of type 2 diabetes (T2D). Here, we used fetal hepatic catheterization to directly measure HGP and substrate flux in a sheep FGR model. We hypothesized that FGR fetuses would have increased hepatic lactate and amino acid uptake to support increased HGP. Indeed, FGR fetuses compared with normal (CON) fetuses had increased HGP and activation of gluconeogenic genes. Unexpectedly, hepatic pyruvate output was increased, while hepatic lactate and gluconeogenic amino acid uptake rates were decreased in FGR liver. Hepatic oxygen consumption and total substrate uptake rates were lower. In FGR liver tissue, metabolite abundance, 13C-metabolite labeling, enzymatic activity, and gene expression supported decreased pyruvate oxidation and increased lactate production. Isolated hepatocytes from FGR fetuses had greater intrinsic capacity for lactate-fueled glucose production. FGR livers also had lower energy (ATP) and redox state (NADH/NAD+ ratio). Thus, reduced hepatic oxidative metabolism may make carbons available for increased HGP, but also produces nutrient and energetic stress in FGR liver. Intrinsic programming of these pathways regulating HGP in the FGR fetus may underlie increased HGP and T2D risk postnatally.
Collapse
Affiliation(s)
- Laura D Brown
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Paul J Rozance
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Dong Wang
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Evren C Eroglu
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Randall B Wilkening
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ashley Solmonson
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Stephanie R Wesolowski
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
33
|
Furukawa F, Aoyagi A, Sano K, Sameshima K, Goto M, Tseng YC, Ikeda D, Lin CC, Uchida K, Okumura SI, Yasumoto K, Jimbo M, Hwang PP. Gluconeogenesis in the extraembryonic yolk syncytial layer of the zebrafish embryo. PNAS NEXUS 2024; 3:pgae125. [PMID: 38585339 PMCID: PMC10997050 DOI: 10.1093/pnasnexus/pgae125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 03/11/2024] [Indexed: 04/09/2024]
Abstract
Yolk-consuming (lecithotrophic) embryos of oviparous animals, such as those of fish, need to make do with the maternally derived yolk. However, in many cases, yolk possesses little carbohydrates and sugars, including glucose, the essential monosaccharide. Interestingly, increases in the glucose content were found in embryos of some teleost fishes; however, the origin of this glucose has been unknown. Unveiling new metabolic strategies in fish embryos has a potential for better aquaculture technologies. In the present study, using zebrafish, we assessed how these embryos obtain the glucose. We employed stable isotope (13C)-labeled substrates and injected them to the zebrafish embryos. Our liquid chromatography-mass spectrometry-based isotope tracking revealed that among all tested substrate, glutamate was most actively metabolized to produce glucose in the zebrafish embryos. Expression analysis for gluconeogenic genes found that many of these were expressed in the yolk syncytial layer (YSL), an extraembryonic tissue found in teleost fishes. Generation 0 (G0) knockout of pck2, a gene encoding the key enzyme for gluconeogenesis from Krebs cycle intermediates, reduced gluconeogenesis from glutamate, suggesting that this gene is responsible for gluconeogenesis from glutamate in the zebrafish embryos. These results showed that teleost YSL undergoes gluconeogenesis, likely contributing to the glucose supplementation to the embryos with limited glucose source. Since many other animal lineages lack YSL, further comparative analysis will be interesting.
Collapse
Affiliation(s)
- Fumiya Furukawa
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2, Nankang, Taipei 11529, Taiwan ROC
| | - Akihiro Aoyagi
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Kaori Sano
- Department of Chemistry, Faculty of Science, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Keita Sameshima
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Miku Goto
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Yung-Che Tseng
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2, Nankang, Taipei 11529, Taiwan ROC
| | - Daisuke Ikeda
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Ching-Chun Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2, Nankang, Taipei 11529, Taiwan ROC
| | - Katsuhisa Uchida
- Department of Marine Biology and Environmental Sciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen Kibanadai-Nishi, Miyazaki 889-2192, Japan
| | - Sei-ichi Okumura
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Ko Yasumoto
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Mitsuru Jimbo
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2, Nankang, Taipei 11529, Taiwan ROC
| |
Collapse
|
34
|
Martino MR, Habibi M, Ferguson D, Brookheart RT, Thyfault JP, Meyer GA, Lantier L, Hughey CC, Finck BN. Disruption of hepatic mitochondrial pyruvate and amino acid metabolism impairs gluconeogenesis and endurance exercise capacity in mice. Am J Physiol Endocrinol Metab 2024; 326:E515-E527. [PMID: 38353639 PMCID: PMC11193532 DOI: 10.1152/ajpendo.00258.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/25/2024] [Accepted: 02/08/2024] [Indexed: 02/23/2024]
Abstract
Exercise robustly increases the glucose demands of skeletal muscle. This demand is met by not only muscle glycogenolysis but also accelerated liver glucose production from hepatic glycogenolysis and gluconeogenesis to fuel mechanical work and prevent hypoglycemia during exercise. Hepatic gluconeogenesis during exercise is dependent on highly coordinated responses within and between muscle and liver. Specifically, exercise increases the rate at which gluconeogenic precursors such as pyruvate/lactate or amino acids are delivered from muscle to the liver, extracted by the liver, and channeled into glucose. Herein, we examined the effects of interrupting hepatic gluconeogenic efficiency and capacity on exercise performance by deleting mitochondrial pyruvate carrier 2 (MPC2) and/or alanine transaminase 2 (ALT2) in the liver of mice. We found that deletion of MPC2 or ALT2 alone did not significantly affect time to exhaustion or postexercise glucose concentrations in treadmill exercise tests, but mice lacking both MPC2 and ALT2 in hepatocytes (double knockout, DKO) reached exhaustion faster and exhibited lower circulating glucose during and after exercise. Use of 2H/1³C metabolic flux analyses demonstrated that DKO mice exhibited lower endogenous glucose production owing to decreased glycogenolysis and gluconeogenesis at rest and during exercise. Decreased gluconeogenesis was accompanied by lower anaplerotic, cataplerotic, and TCA cycle fluxes. Collectively, these findings demonstrate that the transition of the liver to the gluconeogenic mode is critical for preventing hypoglycemia and sustaining performance during exercise. The results also illustrate the need for interorgan cross talk during exercise as described by the Cahill and Cori cycles.NEW & NOTEWORTHY Martino and colleagues examined the effects of inhibiting hepatic gluconeogenesis on exercise performance and systemic metabolism during treadmill exercise in mice. Combined inhibition of gluconeogenesis from lactate/pyruvate and alanine impaired exercise endurance and led to hypoglycemia during and after exercise. In contrast, suppressing either pyruvate-mediated or alanine-mediated gluconeogenesis alone had no effect on these parameters. These findings provide new insight into the molecular nodes that coordinate the metabolic responses of muscle and liver during exercise.
Collapse
Affiliation(s)
- Michael R Martino
- Division of Nutritional Sciences and Obesity Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Mohammad Habibi
- Division of Nutritional Sciences and Obesity Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Daniel Ferguson
- Division of Nutritional Sciences and Obesity Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Rita T Brookheart
- Division of Nutritional Sciences and Obesity Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - John P Thyfault
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Missouri, United States
| | - Gretchen A Meyer
- Department of Medicine, Program in Physical Therapy, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Curtis C Hughey
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
| | - Brian N Finck
- Division of Nutritional Sciences and Obesity Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
35
|
Ren Y, Sun J, Mao X. Quality changes in gazami crab (Portunus trituberculatus) during refrigeration. Food Chem 2024; 437:137942. [PMID: 37951080 DOI: 10.1016/j.foodchem.2023.137942] [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: 06/25/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/13/2023]
Abstract
Gazami crab (Portunus trituberculatus) is prone to spoilage during storage and transportation. More research is needed to determine how to reliably show its freshness and explain the mechanism of quality deterioration. We hypothesized that proteins extracted from crabs can be biomarkers to detect crab muscle quality changes. This work used physicochemical and proteomic approaches to investigate protein biomarkers and molecular mechanisms driving changes in gazami crab muscle quality after long-term refrigeration. It was shown that 66 differentially abundant proteins (DAPs) were closely associated with pH and texture and can be used as biomarkers to assess crab muscle freshness. According to bioinformatics studies, ribosomes and autophagy were significant mechanisms in crab rotting. These findings provided new concepts and a theoretical foundation for evaluating the freshness of refrigerated gazami crab and help uncover the molecular mechanism of its quality deterioration.
Collapse
Affiliation(s)
- Yanmei Ren
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, PR China; Qingdao Key Laboratory of Food Biotechnology, Qingdao 266404, PR China; Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao 266404, PR China
| | - Jianan Sun
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, PR China; Qingdao Key Laboratory of Food Biotechnology, Qingdao 266404, PR China; Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao 266404, PR China.
| | - Xiangzhao Mao
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, PR China; Qingdao Key Laboratory of Food Biotechnology, Qingdao 266404, PR China; Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao 266404, PR China.
| |
Collapse
|
36
|
Popova D, Sun J, Chow HM, Hart RP. A critical review of ethanol effects on neuronal firing: A metabolic perspective. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:450-458. [PMID: 38217065 PMCID: PMC10966925 DOI: 10.1111/acer.15266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
Ethanol metabolism is relatively understudied in neurons, even though changes in neuronal metabolism are known to affect their activity. Recent work demonstrates that ethanol is preferentially metabolized over glucose as a source of carbon and energy, and it reprograms neurons to a state of reduced energy potential and diminished capacity to utilize glucose once ethanol is exhausted. Ethanol intake has been associated with changes in neuronal firing and specific brain activity (EEG) patterns have been linked with risk for alcohol use disorder (AUD). Furthermore, a haplotype of the inwardly rectifying potassium channel subunit, GIRK2, which plays a critical role in regulating excitability of neurons, has been linked with AUD and shown to be directly regulated by ethanol. At the same time, overexpression of GIRK2 prevents ethanol-induced metabolic changes. Based on the available evidence, we conclude that the mechanisms underlying the effects of ethanol on neuronal metabolism are a novel target for developing therapies for AUD.
Collapse
Affiliation(s)
- Dina Popova
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway NJ USA
- Present address: Neuroscience Institute, NYU Langone Grossman School of Medicine, New York, NY USA
| | - Jacquelyne Sun
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Hei-Man Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Ronald P. Hart
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway NJ USA
| |
Collapse
|
37
|
Fu B, Lou Y, Lu X, Wu Z, Ni J, Jin C, Wu P, Xu C. tRF-1:30-Gly-CCC-3 inhibits thyroid cancer via binding to PC and modulating metabolic reprogramming. Life Sci Alliance 2024; 7:e202302285. [PMID: 38081642 PMCID: PMC10713435 DOI: 10.26508/lsa.202302285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
tRFs and tiRNAs (tRNA-derived fragments) are an emerging class of small noncoding RNAs produced by the precise shearing of tRNAs in response to specific stimuli. They have been reported to regulate the pathological processes of numerous human cancers. However, the biofunction of tRFs and tiRNAs in the development and progression of papillary thyroid cancer (PTC) has not been reported yet. In this study, we aimed to explore the biological roles of tRFs and tiRNAs in PTC and discovered that a novel 5'tRNA-derived fragment called tRF-1:30-Gly-CCC-3 (tRF-30) was markedly down-regulated in PTC tissues and cell lines. Functionally, tRF-30 inhibited the proliferation and invasion of PTC cells. Mechanistically, tRF-30 directly bound to the biotin-dependent enzyme pyruvate carboxylase (PC), downregulated its protein level, interfered with the TCA cycle intermediate anaplerosis, and thus affected metabolic reprogramming and PTC progression. These findings revealed a novel regulatory mechanism for tRFs and a potential therapeutic target for PTC.
Collapse
Affiliation(s)
- Bifei Fu
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - YuMing Lou
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiaofeng Lu
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Zhaolin Wu
- Department of Anaesthesiology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Junjie Ni
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Cong Jin
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Pu Wu
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Chaoyang Xu
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
38
|
Liang QH, Li QR, Chen Z, Lv LJ, Lin Y, Jiang HL, Wang KX, Xiao MY, Kang NX, Tu PF, Ji SL, Deng KJ, Gao HW, Zhang L, Li K, Ge F, Xu GQ, Yang SL, Liu YL, Xu QM. Anemoside B4, a new pyruvate carboxylase inhibitor, alleviates colitis by reprogramming macrophage function. Inflamm Res 2024; 73:345-362. [PMID: 38157008 DOI: 10.1007/s00011-023-01840-x] [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: 08/08/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024] Open
Abstract
OBJECTIVES Colitis is a global disease usually accompanied by intestinal epithelial damage and intestinal inflammation, and an increasing number of studies have found natural products to be highly effective in treating colitis. Anemoside B4 (AB4), an abundant saponin isolated from Pulsatilla chinensis (Bunge), which was found to have strong anti-inflammatory activity. However, the exact molecular mechanisms and direct targets of AB4 in the treatment of colitis remain to be discovered. METHODS The anti-inflammatory activities of AB4 were verified in LPS-induced cell models and 2, 4, 6-trinitrobenzene sulfonic (TNBS) or dextran sulfate sodium (DSS)-induced colitis mice and rat models. The molecular target of AB4 was identified by affinity chromatography analysis using chemical probes derived from AB4. Experiments including proteomics, molecular docking, biotin pull-down, surface plasmon resonance (SPR), and cellular thermal shift assay (CETSA) were used to confirm the binding of AB4 to its molecular target. Overexpression of pyruvate carboxylase (PC) and PC agonist were used to study the effects of PC on the anti-inflammatory and metabolic regulation of AB4 in vitro and in vivo. RESULTS AB4 not only significantly inhibited LPS-induced NF-κB activation and increased ROS levels in THP-1 cells, but also suppressed TNBS/DSS-induced colonic inflammation in mice and rats. The molecular target of AB4 was identified as PC, a key enzyme related to fatty acid, amino acid and tricarboxylic acid (TCA) cycle. We next demonstrated that AB4 specifically bound to the His879 site of PC and altered the protein's spatial conformation, thereby affecting the enzymatic activity of PC. LPS activated NF-κB pathway and increased PC activity, which caused metabolic reprogramming, while AB4 reversed this phenomenon by inhibiting the PC activity. In vivo studies showed that diisopropylamine dichloroacetate (DADA), a PC agonist, eliminated the therapeutic effects of AB4 by changing the metabolic rearrangement of intestinal tissues in colitis mice. CONCLUSION We identified PC as a direct cellular target of AB4 in the modulation of inflammation, especially colitis. Moreover, PC/pyruvate metabolism/NF-κB is crucial for LPS-driven inflammation and oxidative stress. These findings shed more light on the possibilities of PC as a potential new target for treating colitis.
Collapse
Affiliation(s)
- Qing-Hua Liang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Qiu-Rong Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Zhong Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Li-Juan Lv
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yu Lin
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hong-Lv Jiang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Ke-Xin Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Ming-Yue Xiao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Nai-Xin Kang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Shi-Liang Ji
- Department of Pharmacy, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, 215163, Jiangsu, China
| | - Ke-Jun Deng
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
| | - Hong-Wei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, Guangxi, China
- Guangxi Xinhai Pharmaceutical Technology Co.,Ltd, , Liuzhou, 545025, Guangxi, China
| | - Li Zhang
- Instrumental Analysis Center, Shanghai JiaoTong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Kun Li
- Hai'an Traditional Chinese Medicine Hospital, Nantong, 226600, Jiangsu, China
| | - Fei Ge
- Hai'an Traditional Chinese Medicine Hospital, Nantong, 226600, Jiangsu, China
| | - Guo-Qiang Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Shi-Lin Yang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, Guangxi, China
- Guangxi Xinhai Pharmaceutical Technology Co.,Ltd, , Liuzhou, 545025, Guangxi, China
| | - Yan-Li Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
| | - Qiong-Ming Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
- Guangxi Xinhai Pharmaceutical Technology Co.,Ltd, , Liuzhou, 545025, Guangxi, China.
| |
Collapse
|
39
|
Alemany M. The Metabolic Syndrome, a Human Disease. Int J Mol Sci 2024; 25:2251. [PMID: 38396928 PMCID: PMC10888680 DOI: 10.3390/ijms25042251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
This review focuses on the question of metabolic syndrome (MS) being a complex, but essentially monophyletic, galaxy of associated diseases/disorders, or just a syndrome of related but rather independent pathologies. The human nature of MS (its exceptionality in Nature and its close interdependence with human action and evolution) is presented and discussed. The text also describes the close interdependence of its components, with special emphasis on the description of their interrelations (including their syndromic development and recruitment), as well as their consequences upon energy handling and partition. The main theories on MS's origin and development are presented in relation to hepatic steatosis, type 2 diabetes, and obesity, but encompass most of the MS components described so far. The differential effects of sex and its biological consequences are considered under the light of human social needs and evolution, which are also directly related to MS epidemiology, severity, and relations with senescence. The triggering and maintenance factors of MS are discussed, with especial emphasis on inflammation, a complex process affecting different levels of organization and which is a critical element for MS development. Inflammation is also related to the operation of connective tissue (including the adipose organ) and the widely studied and acknowledged influence of diet. The role of diet composition, including the transcendence of the anaplerotic maintenance of the Krebs cycle from dietary amino acid supply (and its timing), is developed in the context of testosterone and β-estradiol control of the insulin-glycaemia hepatic core system of carbohydrate-triacylglycerol energy handling. The high probability of MS acting as a unique complex biological control system (essentially monophyletic) is presented, together with additional perspectives/considerations on the treatment of this 'very' human disease.
Collapse
Affiliation(s)
- Marià Alemany
- Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
40
|
Yang Q, Wu Y, Liu W, Ou X, Zhang W, Wang J, Chang Y, Wang F, Gao M, Liu S. Zonated iron deposition in the periportal zone of the liver is associated with selectively enhanced lipid synthesis. Liver Int 2024; 44:589-602. [PMID: 38082474 DOI: 10.1111/liv.15807] [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: 02/28/2023] [Revised: 10/28/2023] [Accepted: 11/17/2023] [Indexed: 01/31/2024]
Abstract
BACKGROUND AND AIMS Disorders in liver lipid metabolism have been implicated in a range of metabolic conditions, including fatty liver and liver cancer. Altered lipid distribution within the liver, shifting from the pericentral to the periportal zone under pathological circumstances, has been observed; however, the underlying mechanism remains elusive. Iron, an essential metal, exhibits a zonal distribution in the liver similar to that of lipids. Nevertheless, the precise relationship between iron and lipid distribution, especially in the pericentral and periportal zones, remains poorly understood. METHODS We conducted comprehensive in vitro and in vivo experiments, combining with in situ analysis and RNA sequencing, aiming for a detailed exploration of the causal relationship between iron accumulation and lipid metabolism. RESULTS Our research suggests that iron overload can disrupt the normal distribution of lipids within the liver, particularly in the periportal zone. Through meticulous gene expression profiling in both the pericentral and periportal zones, we identified pyruvate carboxylase (PC) as a pivotal regulator in iron overload-induced lipid accumulation. Additionally, we revealed that the activation of cyclic adenosine monophosphate response element binding protein (CREB) was indispensable for Pc gene expression when in response to iron overload. CONCLUSIONS In summary, our investigation unveils the crucial involvement of iron overload in fostering hepatic lipid accumulation in the periportal zone, at least partly mediated by the modulation of Pc expression. These insights offer new perspectives for understanding the pathogenesis of fatty liver diseases and their progression.
Collapse
Affiliation(s)
- Qiuyuan Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yue Wu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wei Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaojuan Ou
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Wei Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Jianning Wang
- The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Yanzhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, China
| | - Fudi Wang
- The Fourth Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Gao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
41
|
Zhao Y, Li C, Zhou S, He Y, Wang Y, Zhang Y, Wen L. Enhanced glucose utilization of skeletal muscle after 4 weeks of intermittent hypoxia in a mouse model of type 2 diabetes. PLoS One 2024; 19:e0296815. [PMID: 38271325 PMCID: PMC10810429 DOI: 10.1371/journal.pone.0296815] [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: 08/12/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Intermittent hypoxia intervention (IHI) has been shown to reduces blood glucose and improves insulin resistance in type 2 diabetes (T2D) and has been suggested as a complementary or alternative intervention to exercise for individuals with limited mobility. Previous research on IHI has assessed cellular glucose uptake rather than utilization. The purpose of this study was to determine the effect of a 4-week IHI, with or without an aerobic exercise, on skeletal muscle glucose utilization as indicated by the changes in pyruvate, lactate, NAD+, and NADH, using a mouse model of diet-induced T2D. In addition, the effects of one exposure to hypoxia (acute) and of a 4-week IHI (chronic) were compared to explore their relationship. METHODS C57BL/6J mice were randomly assigned to normal control and high-fat-diet groups, and the mice that developed diet-induced diabetes were assigned to diabetes control, and intervention groups with 1 hour (acute) or 4 weeks (1 hour/day, 6 days/week) exposure to a hypoxic envrionment (0.15 FiO2), exercise (treadmill run) in normoxia, and exercise in hypoxia, respectively, with N = 7 in each group. The effects of the interventions on concentrations of fasting blood glucose, muscle glucose, GLUT4, lactate, pyruvate, nicotinamide adenine dinucleotide (NAD+), and NADH were measured, and statistically compared between the groups. RESULTS Compared with diabetes control group, the mice treated in the hypoxic environment for 4 weeks showed a significantly higher pyruvate levels and lower lactate/pyruvate ratios in the quadriceps muscle, and the mice exposed to hypoxia without or with aerobic exercise for either for 4 weeks or just 1 hour showed higher NAD+ levels and lower NADH/NAD+ ratios. CONCLUSIONS Exposure to moderate hypoxia for either one bout or 4 weeks significantly increased the body's mitochondrial NAD cyclethe in diabetic mice even in the absence of aerobic exercise. The hypoxia and exercise interventions exhibited synergistic effects on glycolysis. These findings provide mechanistic insights into the effects of IHI in respect of the management of hyperglycemia.
Collapse
Affiliation(s)
- Yuqi Zhao
- School of Social Sports and Health Sciences, Tianjin University of Sport, Tianjin, China
- School of Exercise and Health, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Chaoqun Li
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Shi Zhou
- Faculty of Health, Southern Cross University, Lismore, Australia
| | - Youyu He
- School of Social Sports and Health Sciences, Tianjin University of Sport, Tianjin, China
| | - Yun Wang
- Faculty of Health, Southern Cross University, Lismore, Australia
| | - Yuan Zhang
- Faculty of Health, Southern Cross University, Lismore, Australia
| | - Li Wen
- School of Social Sports and Health Sciences, Tianjin University of Sport, Tianjin, China
- School of Exercise and Health, Nanjing Sport Institute, Nanjing, Jiangsu, China
| |
Collapse
|
42
|
Sukjoi W, Young C, Acland M, Siritutsoontorn S, Roytrakul S, Klingler-Hoffmann M, Hoffmann P, Jitrapakdee S. Proteomic analysis of holocarboxylase synthetase deficient-MDA-MB-231 breast cancer cells revealed the biochemical changes associated with cell death, impaired growth signaling, and metabolism. Front Mol Biosci 2024; 10:1250423. [PMID: 38283944 PMCID: PMC10812114 DOI: 10.3389/fmolb.2023.1250423] [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: 06/30/2023] [Accepted: 12/27/2023] [Indexed: 01/30/2024] Open
Abstract
We have previously shown that the holocarboxylase synthetase (HLCS) is overexpressed in breast cancer tissue of patients, and silencing of its expression in triple-negative cancer cell line inhibits growth and migration. Here we investigated the global biochemical changes associated with HLCS knockdown in MDA-MB-231 cells to discern the pathways that involve HLCS. Proteomic analysis of two independent HLCS knockdown cell lines identified 347 differentially expressed proteins (DEPs) whose expression change > 2-fold (p < 0.05) relative to the control cell line. GO enrichment analysis showed that these DEPs were mainly associated with the cellular process such as cellular metabolic process, cellular response to stimulus, and cellular component organization or biogenesis, metabolic process, biological regulation, response to stimuli, localization, and signaling. Among the 347 identified DEPs, 64 proteins were commonly found in both HLCS knockdown clones, confirming their authenticity. Validation of some of these DEPs by Western blot analysis showed that plasminogen activator inhibitor type 2 (SerpinB2) and interstitial collagenase (MMP1) were approximately 90% decreased in HLCS knockdown cells, consistent with a 50%-60% decrease in invasion ability of knockdown cells. Notably, argininosuccinate synthase 1 (ASS1), one of the enzymes in the urea cycle, showed approximately a 10-fold increase in the knockdown cells, suggesting the crucial role of HLCS in supporting the urea cycle in the triple-negative cancer cell line. Collectively, our proteomic data provide biochemical insights into how suppression of HLCS expression perturbs global changes in cellular processes and metabolic pathways, impairing cell growth and invasion.
Collapse
Affiliation(s)
- Witchuda Sukjoi
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Clifford Young
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Mitchell Acland
- Adelaide Proteomics Centre, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | | | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Agency, Pathumthani, Thailand
| | | | - Peter Hoffmann
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sarawut Jitrapakdee
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
43
|
Yu C, Wang D, Shen C, Luo Z, Zhang H, Zhang J, Xu W, Xu J. Remodeling of Hepatic Glucose Metabolism in Response to Early Weaning in Piglets. Animals (Basel) 2024; 14:190. [PMID: 38254359 PMCID: PMC10812452 DOI: 10.3390/ani14020190] [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: 11/16/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
This study aimed to investigate the dynamic changes in hepatic glucose metabolism in response to early weaning. A total of 60 piglets were randomly selected and weaned at 21 days old. Six piglets were slaughtered on the weaning day (d0) and at 1 (d1), 4 (d4), 7 (d7), and 14 (d14) days postweaning. The results illustrated that body weight significantly increased from d4 to d14 (p < 0.001). Serum glucose fell sharply after weaning and then remained at a low level from d1 to d14 (p < 0.001). Serum insulin decreased from d4 (p < 0.001), which caused hepatic glycogen to be broken down (p = 0.007). The glucose-6-phosphatase activity increased from d0 to d4 and then decreased from d4 to d14 (p = 0.039). The pyruvate carboxylase activity presented a significant sustained increase from d0 to d14 (p < 0.001). The succinate (p = 0.006) and oxaloacetate (p = 0.003) content on d4 was lower than that on d0. The succinate dehydrogenase activity (p = 0.008) and ATP (p = 0.016) production decreased significantly on d4 compared to that on d0. Taken together, these findings reveal the dynamic changes of metabolites and enzymes related to hepatic glycometabolism and the TCA (tricarboxylic acid) cycle in piglets after weaning. Our findings enrich weaning stress theory and might provide a reference for dietary intervention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jianxiong Xu
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (C.Y.)
| |
Collapse
|
44
|
Yiew NK, Deja S, Ferguson D, Cho K, Jarasvaraparn C, Jacome-Sosa M, Lutkewitte AJ, Mukherjee S, Fu X, Singer JM, Patti GJ, Burgess SC, Finck BN. Effects of hepatic mitochondrial pyruvate carrier deficiency on de novo lipogenesis and gluconeogenesis in mice. iScience 2023; 26:108196. [PMID: 37942005 PMCID: PMC10628847 DOI: 10.1016/j.isci.2023.108196] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/31/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
The liver coordinates the systemic response to nutrient deprivation and availability by producing glucose from gluconeogenesis during fasting and synthesizing lipids via de novo lipogenesis (DNL) when carbohydrates are abundant. Mitochondrial pyruvate metabolism is thought to play important roles in both gluconeogenesis and DNL. We examined the effects of hepatocyte-specific mitochondrial pyruvate carrier (MPC) deletion on the fasting-refeeding response. Rates of DNL during refeeding were impaired by hepatocyte MPC deletion, but this did not reduce intrahepatic lipid content. During fasting, glycerol is converted to glucose by two pathways; a direct cytosolic pathway and an indirect mitochondrial pathway requiring the MPC. Hepatocyte MPC deletion reduced the incorporation of 13C-glycerol into TCA cycle metabolites, but not into new glucose. Furthermore, suppression of glycerol and alanine metabolism did not affect glucose concentrations in fasted hepatocyte-specific MPC-deficient mice, suggesting multiple layers of redundancy in glycemic control in mice.
Collapse
Affiliation(s)
- Nicole K.H. Yiew
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Stanislaw Deja
- Center for Human Nutrition, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Daniel Ferguson
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kevin Cho
- Department of Chemistry, Siteman Cancer Center, Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Chaowapong Jarasvaraparn
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Miriam Jacome-Sosa
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Andrew J. Lutkewitte
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Sandip Mukherjee
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Xiaorong Fu
- Center for Human Nutrition, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Jason M. Singer
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Gary J. Patti
- Department of Chemistry, Siteman Cancer Center, Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Shawn C. Burgess
- Center for Human Nutrition, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Brian N. Finck
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
45
|
Che Z, Zhou Z, Li SQ, Gao L, Xiao J, Wong NK. ROS/RNS as molecular signatures of chronic liver diseases. Trends Mol Med 2023; 29:951-967. [PMID: 37704494 DOI: 10.1016/j.molmed.2023.08.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/11/2023] [Accepted: 08/07/2023] [Indexed: 09/15/2023]
Abstract
The liver can succumb to oxidant damage during the development of chronic liver diseases. Despite their physiological relevance to hepatic homeostasis, excessive reactive oxygen/nitrogen species (ROS/RNS) production under pathological conditions is detrimental to all liver constituents. Chronic oxidative stress coupled to unresolved inflammation sets in motion the activation of profibrogenic hepatic stellate cells (HSCs) and later pathogenesis of liver fibrosis, cirrhosis, and liver cancer. The liver antioxidant and repair systems, along with autophagic and ferroptotic machineries, are implicated in the onset and trajectory of disease development. In this review, we discuss the ROS/RNS-related mechanisms underlying liver fibrosis of distinct etiologies and highlight preclinical and clinical trials of antifibrotic therapies premised on remediating oxidative/nitrosative stress in hepatocytes or targeting HSC activation.
Collapse
Affiliation(s)
- Zhaodi Che
- Clinical Research Institute, Institute of Obesity and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
| | - Ziyuan Zhou
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; Clinical Pharmacology Section, Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Si-Qi Li
- Clinical Pharmacology Section, Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Jia Xiao
- Clinical Research Institute, Institute of Obesity and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou 510000, China; Shandong Provincial Key Laboratory for Clinical Research of Liver Diseases, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao 266001, China.
| | - Nai-Kei Wong
- Clinical Pharmacology Section, Department of Pharmacology, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
46
|
Yiew NKH, Vazquez JH, Martino MR, Kennon-McGill S, Price JR, Allard FD, Yee EU, Layman AJ, James LP, McCommis KS, Finck BN, McGill MR. Hepatic pyruvate and alanine metabolism are critical and complementary for maintenance of antioxidant capacity and resistance to oxidative insult. Mol Metab 2023; 77:101808. [PMID: 37716594 PMCID: PMC10561123 DOI: 10.1016/j.molmet.2023.101808] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023] Open
Abstract
OBJECTIVE Mitochondrial pyruvate is a critical intermediary metabolite in gluconeogenesis, lipogenesis, and NADH production. As a result, the mitochondrial pyruvate carrier (MPC) complex has emerged as a promising therapeutic target in metabolic diseases. Clinical trials are currently underway. However, recent in vitro data indicate that MPC inhibition diverts glutamine/glutamate away from glutathione synthesis and toward glutaminolysis to compensate for loss of pyruvate oxidation, possibly sensitizing cells to oxidative insult. Here, we explored this in vivo using the clinically relevant acetaminophen (APAP) overdose model of acute liver injury, which is driven by oxidative stress. METHODS We used pharmacological and genetic approaches to inhibit MPC2 and alanine aminotransferase 2 (ALT2), individually and concomitantly, in mice and cell culture models and determined the effects on APAP hepatotoxicity. RESULTS We found that MPC inhibition sensitizes the liver to APAP-induced injury in vivo only with concomitant loss of alanine aminotransferase 2 (ALT2). Pharmacological and genetic manipulation of neither MPC2 nor ALT2 alone affected APAP toxicity, but liver-specific double knockout (DKO) significantly worsened APAP-induced liver damage. Further investigation indicated that DKO impaired glutathione synthesis and increased urea cycle flux, consistent with increased glutaminolysis, and these results were reproducible in vitro. Finally, induction of ALT2 and post-treatment with dichloroacetate both reduced APAP-induced liver injury, suggesting new therapeutic avenues. CONCLUSIONS Increased susceptibility to APAP toxicity requires loss of both the MPC and ALT2 in vivo, indicating that MPC inhibition alone is insufficient to disrupt redox balance. Furthermore, the results from ALT2 induction and dichloroacetate in the APAP model suggest new metabolic approaches to the treatment of liver damage.
Collapse
Affiliation(s)
- Nicole K H Yiew
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Joel H Vazquez
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Michael R Martino
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Stefanie Kennon-McGill
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jake R Price
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Felicia D Allard
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Eric U Yee
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Alexander J Layman
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Laura P James
- Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kyle S McCommis
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Brian N Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mitchell R McGill
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
47
|
Shrivastava S, Sharma A, Saxena N, Bhamra R, Kumar S. Addressing the preventive and therapeutic perspective of berberine against diabetes. Heliyon 2023; 9:e21233. [PMID: 38027723 PMCID: PMC10663750 DOI: 10.1016/j.heliyon.2023.e21233] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/20/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Diabetes has emerged as one the leading detrimental factors for human life expectancy worldwide. The disease is mainly considered as outcome of dysregulation in glucose metabolism, resulting in consistent high glucose concentration in blood. At initial stages, the diabetes particularly type 2 diabetes, is manageable by lifestyle interventions such as regular physical activity and diet with less carbohydrates. However, in advance stage, regular intake of external insulin dose and medicines like metformin are recommended. The long-term consumption of metformin is associated with several side effects such as nausea, vomiting, diarrhoea, lectic acidosis etc., In this scenario, several plant-based medicines have shown promising potential for the prevention and treatment of diabetes. Berberine is the bioactive compound present in the different plant parts of berberis family. Biochemical studies have shown that berberine improve insulin sensitivity and insulin secretion. Additionally, berberine induces glucose metabolism by activating AMPK signaling and inhibition of inflammation. A series of studies have demonstrated the antidiabetic potential of berberine at in vitro, pre-clinical and clinical trials. This review provides comprehensive details of preventive and therapeutic potential of berberine against diabetes.
Collapse
Affiliation(s)
- Suyesh Shrivastava
- ICMR-National Institute of Research in Tribal Health, Nagpur Road, Jabalpur-482003, India
| | - Anamika Sharma
- National Institute of Pharmaceutical and Education and Research 500037, Hyderabad, India
| | - Nishant Saxena
- ICMR-National Institute of Research in Tribal Health, Nagpur Road, Jabalpur-482003, India
| | - Rashmi Bhamra
- Global Research Institute of Pharmacy, Radour-135133, Haryana, India
| | - Sandeep Kumar
- ICMR-National Institute of Research in Tribal Health, Nagpur Road, Jabalpur-482003, India
| |
Collapse
|
48
|
Onodera T, Wang MY, Rutkowski JM, Deja S, Chen S, Balzer MS, Kim DS, Sun X, An YA, Field BC, Lee C, Matsuo EI, Mizerska M, Sanjana I, Fujiwara N, Kusminski CM, Gordillo R, Gautron L, Marciano DK, Hu MC, Burgess SC, Susztak K, Moe OW, Scherer PE. Endogenous renal adiponectin drives gluconeogenesis through enhancing pyruvate and fatty acid utilization. Nat Commun 2023; 14:6531. [PMID: 37848446 PMCID: PMC10582045 DOI: 10.1038/s41467-023-42188-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 10/03/2023] [Indexed: 10/19/2023] Open
Abstract
Adiponectin is a secretory protein, primarily produced in adipocytes. However, low but detectable expression of adiponectin can be observed in cell types beyond adipocytes, particularly in kidney tubular cells, but its local renal role is unknown. We assessed the impact of renal adiponectin by utilizing male inducible kidney tubular cell-specific adiponectin overexpression or knockout mice. Kidney-specific adiponectin overexpression induces a doubling of phosphoenolpyruvate carboxylase expression and enhanced pyruvate-mediated glucose production, tricarboxylic acid cycle intermediates and an upregulation of fatty acid oxidation (FAO). Inhibition of FAO reduces the adiponectin-induced enhancement of glucose production, highlighting the role of FAO in the induction of renal gluconeogenesis. In contrast, mice lacking adiponectin in the kidney exhibit enhanced glucose tolerance, lower utilization and greater accumulation of lipid species. Hence, renal adiponectin is an inducer of gluconeogenesis by driving enhanced local FAO and further underlines the important systemic contribution of renal gluconeogenesis.
Collapse
Affiliation(s)
- Toshiharu Onodera
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, US
| | - May-Yun Wang
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, US
| | - Joseph M Rutkowski
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Stanislaw Deja
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, US
| | - Shiuhwei Chen
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, US
| | - Michael S Balzer
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Nephrology and Medical Intensive Care, Charité, Universitätsmedizin Berlin, 10117, Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, 10117, Berlin, Germany
| | - Dae-Seok Kim
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, US
| | - Xuenan Sun
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, US
| | - Yu A An
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, US
- Department of Anesthesiology, Critical Care and Pain Medicine, UT Health Science Center at Houston, Houston, TX, USA
| | - Bianca C Field
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, US
| | - Charlotte Lee
- Center for Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ei-Ichi Matsuo
- Solutions COE, Analytical & Measuring Instruments Division, Shimadzu Corporation, Kyoto, Japan
| | - Monika Mizerska
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, US
| | - Ina Sanjana
- Solutions COE, Analytical & Measuring Instruments Division, Shimadzu Corporation, Kyoto, Japan
| | - Naoto Fujiwara
- Liver Tumor Translational Research Program, Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Christine M Kusminski
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, US
| | - Ruth Gordillo
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, US
| | - Laurent Gautron
- Center for Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Denise K Marciano
- Departments of Cell Biology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shawn C Burgess
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, US
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, US.
- Departments of Cell Biology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
49
|
Karpale M, Kummu O, Kärkkäinen O, Lehtonen M, Näpänkangas J, Herfurth UM, Braeuning A, Rysä J, Hakkola J. Pregnane X receptor activation remodels glucose metabolism to promote NAFLD development in obese mice. Mol Metab 2023; 76:101779. [PMID: 37467962 PMCID: PMC10415798 DOI: 10.1016/j.molmet.2023.101779] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/14/2023] [Accepted: 07/13/2023] [Indexed: 07/21/2023] Open
Abstract
OBJECTIVE Both obesity and exposure to chemicals may induce non-alcoholic fatty liver disease (NAFLD). Pregnane X Receptor (PXR) is a central target of metabolism disrupting chemicals and disturbs hepatic glucose and lipid metabolism. We hypothesized that the metabolic consequences of PXR activation may be modified by existing obesity and associated metabolic dysfunction. METHODS Wildtype and PXR knockout male mice were fed high-fat diet to induce obesity and metabolic dysfunction. PXR was activated with pregnenolone-16α-carbonitrile. Glucose metabolism, hepatosteatosis, insulin signaling, glucose uptake, liver glycogen, plasma and liver metabolomics, and liver, white adipose tissue, and muscle transcriptomics were investigated. RESULTS PXR activation aggravated obesity-induced liver steatosis by promoting lipogenesis and inhibiting fatty acid disposal. Accordingly, hepatic insulin sensitivity was impaired and circulating alanine aminotransferase level increased. Lipid synthesis was facilitated by increased liver glucose uptake and utilization of glycogen reserves resulting in dissociation of hepatosteatosis and hepatic insulin resistance from the systemic glucose tolerance and insulin sensitivity. Furthermore, glucagon-induced hepatic glucose production was impaired. PXR deficiency did not protect from the metabolic manifestations of obesity, but the liver transcriptomics and metabolomics profiling suggest diminished activation of inflammation and less prominent changes in the overall metabolite profile. CONCLUSIONS Obesity and PXR activation by chemical exposure have a synergistic effect on NAFLD development. To support liver fat accumulation the PXR activation reorganizes glucose metabolism that seemingly improves systemic glucose metabolism. This implies that obese individuals, already predisposed to metabolic diseases, may be more susceptible to harmful metabolic effects of PXR-activating drugs and environmental chemicals.
Collapse
Affiliation(s)
- Mikko Karpale
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Outi Kummu
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Olli Kärkkäinen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Juha Näpänkangas
- Department of Pathology, University of Oulu, Oulu University Hospital, Oulu, Finland
| | - Uta M Herfurth
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, Berlin, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, Berlin, Germany
| | - Jaana Rysä
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Jukka Hakkola
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.
| |
Collapse
|
50
|
Yiew NK, Deja S, Ferguson D, Cho K, Jarasvaraparn C, Jacome-Sosa M, Lutkewitte AJ, Mukherjee S, Fu X, Singer JM, Patti GJ, Burgess SC, Finck BN. Effects of hepatic mitochondrial pyruvate carrier deficiency on de novo lipogenesis and glycerol-mediated gluconeogenesis in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.17.528992. [PMID: 36824879 PMCID: PMC9949129 DOI: 10.1101/2023.02.17.528992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The liver coordinates the systemic response to nutrient deprivation and availability by producing glucose from gluconeogenesis during fasting and synthesizing lipids via de novo lipogenesis (DNL) when carbohydrates are abundant. Mitochondrial pyruvate metabolism is thought to play important roles in both gluconeogenesis and DNL. We examined the effects of hepatocyte-specific mitochondrial pyruvate carrier (MPC) deletion on the fasting-refeeding response. Rates of DNL during refeeding were impaired by liver MPC deletion, but this did not reduce intrahepatic lipid content. During fasting, glycerol is converted to glucose by two pathways; a direct cytosolic pathway essentially reversing glycolysis and an indirect mitochondrial pathway requiring the MPC. MPC deletion reduced the incorporation of 13C-glycerol into TCA cycle metabolites but not into newly synthesized glucose. However, suppression of glycerol metabolism did not affect glucose concentrations in fasted hepatocyte-specific MPC-deficient mice. Thus, glucose production by kidney and intestine may compensate for MPC deficiency in hepatocytes.
Collapse
Affiliation(s)
- Nicole K.H. Yiew
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, MO 63110 USA
| | - Stanislaw Deja
- Center for Human Nutrition, University of Texas Southwestern, Dallas, TX 75390 USA
| | - Daniel Ferguson
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, MO 63110 USA
| | - Kevin Cho
- Department of Chemistry, Siteman Cancer Center, Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, MO 63110 USA
| | - Chaowapong Jarasvaraparn
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, MO 63110 USA
| | - Miriam Jacome-Sosa
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, MO 63110 USA
| | - Andrew J. Lutkewitte
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, MO 63110 USA
| | - Sandip Mukherjee
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, MO 63110 USA
| | - Xiaorong Fu
- Center for Human Nutrition, University of Texas Southwestern, Dallas, TX 75390 USA
| | - Jason M. Singer
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, MO 63110 USA
| | - Gary J. Patti
- Department of Chemistry, Siteman Cancer Center, Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, MO 63110 USA
| | - Shawn C. Burgess
- Center for Human Nutrition, University of Texas Southwestern, Dallas, TX 75390 USA
| | - Brian N. Finck
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, MO 63110 USA
| |
Collapse
|