1
|
Vo ATT, Mouli K, Liopo AV, Lorenzi P, Tan L, Wei B, Martinez SA, McHugh EA, Tour JM, Khan U, Derry PJ, Kent TA. Pleozymes: Pleiotropic Oxidized Carbon Nanozymes Enhance Cellular Metabolic Flexibility. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:2017. [PMID: 39728553 PMCID: PMC11728746 DOI: 10.3390/nano14242017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
Our group has synthesized a pleiotropic synthetic nanozyme redox mediator we term a "pleozyme" that displays multiple enzymatic characteristics, including acting as a superoxide dismutase mimetic, oxidizing NADH to NAD+, and oxidizing H2S to polysulfides and thiosulfate. Benefits have been seen in acute and chronic neurological disease models. The molecule is sourced from coconut-derived activated charcoal that has undergone harsh oxidization with fuming nitric acid, which alters the structure and chemical characteristics, yielding 3-8 nm discs with broad redox potential. Prior work showed pleozymes localize to mitochondria and increase oxidative phosphorylation and glycolysis. Here, we measured cellular NAD+ and NADH levels after pleozyme treatment and observed increased total cellular NADH levels but not total NAD+ levels. A 13C-glucose metabolic flux analysis suggested pleozymes stimulate the generation of pyruvate and lactate glycolytically and from the tricarboxylic acid (TCA) cycle, pointing to malate decarboxylation. Analysis of intracellular fatty acid abundances suggests pleozymes increased fatty acid β-oxidation, with a concomitant increase in succinyl- and acetyl-CoA. Pleozymes increased total ATP, potentially via flexible enhancement of NAD+-dependent catabolic pathways such as glycolysis, fatty acid β-oxidation, and metabolic flux through the TCA cycle. These effects may be favorable for pathologies that compromise metabolism such as brain injury.
Collapse
Affiliation(s)
- Anh T. T. Vo
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
| | - Karthik Mouli
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
| | - Anton V. Liopo
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
| | - Philip Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.L.); (L.T.); (B.W.); (S.A.M.)
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.L.); (L.T.); (B.W.); (S.A.M.)
| | - Bo Wei
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.L.); (L.T.); (B.W.); (S.A.M.)
| | - Sara A. Martinez
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.L.); (L.T.); (B.W.); (S.A.M.)
| | - Emily A. McHugh
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
- Smalley-Curl Institute, Rice University, Houston, TX 77005, USA
| | - James M. Tour
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
- Smalley-Curl Institute, Rice University, Houston, TX 77005, USA
- Rice Advanced Materials Institute, Rice University, Houston, TX 77005, USA
- The NanoCarbon Center, Rice University, Houston, TX 77005, USA
| | - Uffaf Khan
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
| | - Paul J. Derry
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
- School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Thomas A. Kent
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital and Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
2
|
Lv X, Shi B, Ren H, Yang W, Qu L, Obianwuna UE, Lyu X. Identification of Candidate Genes for Sebum Deposition in Pekin Ducks Using Genome-Wide Association Studies. Genes (Basel) 2024; 15:1553. [PMID: 39766820 PMCID: PMC11675992 DOI: 10.3390/genes15121553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Sebum deposition is a vital trait influencing meat quality and production efficiency in Pekin ducks. Providing insights into the genetic basis of fat deposition could help improve breeding strategies aimed at producing high-quality meat ducks. This study aimed to identify the genetic mechanisms and lipid metabolism pathways regulating subcutaneous and intramuscular fat deposition in two Pekin duck strains: Nankou No. 1 and Jingdian. METHODS A total of 72 male ducks, Nankou No. 1 (n = 36) and Jingdian (n = 36), were raised under controlled conditions for 42 days. On days 28, 35, and 42, ducks from each group were selected and slaughtered, and their subcutaneous and liver tissues were collected to analyze lipid enzyme activities. On day 42, additional ducks from each strain were slaughtered and evaluated for carcass performance, as well as intramuscular and sebum yield. Genome-wide association analysis (GWAS) was conducted in the Nankou No. 1 strain. CONCLUSION Our results showed statistically significant differences in intramuscular and subcutaneous fat yield between the two strains, with Nankou No. 1 exhibiting a higher yield than Jingdian (p < 0.05). The GWAS results identified 96 significant single nucleotide polymorphisms (SNPs), associated with sebum deposition. Functional annotation identified ALDH7A1 as a key candidate gene involved in lipid metabolism and fat storage regulation in Pekin ducks, Nankou No. 1 strain. Enzyme activity assays in liver and subcutaneous tissues revealed breed-specific differences in lipid metabolism, aligning with genetic findings. The activities of the lipid enzymes changed over time, suggesting changes in the developmental stages. The results on fat yield and enzymatic activities further align with molecular findings from the GWAS, which identified variations in lipid metabolism pathways. These results highlight genetic markers and biochemical pathways related to fat deposition in Pekin ducks, offering new insights for selective breeding programs aimed at optimizing fat content in meat production. Further research is needed to clarify the specific role of ALDH7A1 in lipid metabolism and its potential to enhance fat deposition traits in poultry.
Collapse
Affiliation(s)
- Xueze Lv
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (X.L.); (L.Q.)
- Product Testing Department, Beijing General Animal Husbandry Station, Beijing 100107, China;
| | - Bozhi Shi
- Instit for the Control of Biological Products, National Institutes for Food and Drug Control, Beijing 102629, China;
| | - Haiyuan Ren
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712199, China;
| | - Weifang Yang
- Product Testing Department, Beijing General Animal Husbandry Station, Beijing 100107, China;
| | - Lujiang Qu
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (X.L.); (L.Q.)
| | | | - Xueqi Lyu
- Emergency Department, Shenzhen New Frontier United Family Healthcare, Shenzhen 518038, China
| |
Collapse
|
3
|
Zhang S, Zhang S, Xia B, Li X, Jiang H, Feng S, Xiang Y, Qiu Y, Zhou S, Luo P. PRMT1-mediated methylation of ME2 promotes hepatocellular carcinoma growth by inhibiting ubiquitination. Cell Death Dis 2024; 15:814. [PMID: 39528487 PMCID: PMC11555414 DOI: 10.1038/s41419-024-07219-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
The mitochondrial malic enzyme 2 (ME2), which is frequently elevated during carcinogenesis and may be a target for cancer therapy, catalyzes the conversion of malate to pyruvate. The processes controlling ME2 activity, however, remain largely unclear. In this work, we show that human hepatocellular carcinoma (HCC) tissues contain high levels of ME2 and that the methylation of ME2 stimulates the growth and migration of HCC cells. Furthermore, we observed that ME2 interacts with protein arginine methyltransferase 1 (PRMT1) and that ME2 enzymatic activity is activated by mutation of ME2 at lysine 67. Mitochondrial respiration was markedly increased by activated ME2, which promoted cell division and carcinogenesis. Furthermore, a negative prognosis for patients was strongly linked with the expression levels of PRMT1 and ME2 R67K in HCC tissues. These findings imply that hepatocellular carcinoma growth is aided by PRMT1-mediated ME2 methylation, that is an essential signaling event that cancer cells need to continue mitochondrial respiration.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, 561113, Guiyang, Guizhou, P.R. China
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
| | - Shuling Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, 561113, Guiyang, Guizhou, P.R. China
| | - Baijuan Xia
- School of Basic Medical Sciences, Guizhou Medical University, 561113, Guiyang, Guizhou, P.R. China
| | - Xueying Li
- Department of Radiology, Guiyang Public Health Clinical Center, No.6 Daying Road, Yunyan District, 550001, Guiyang, Guizhou, P.R. China
| | - Hongyu Jiang
- Department of Cancer Research Laboratory, The Affiliated Cancer Hospital of Guizhou Medical University, 550000, Guiyang, Guizhou, P.R. China
| | - Su Feng
- Department of Cancer Research Laboratory, The Affiliated Cancer Hospital of Guizhou Medical University, 550000, Guiyang, Guizhou, P.R. China
| | - Yang Xiang
- School of Basic Medical Sciences, Guizhou Medical University, 561113, Guiyang, Guizhou, P.R. China
| | - Ya Qiu
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, 561113, Guiyang, Guizhou, P.R. China
| | - Shi Zhou
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China.
| | - Peng Luo
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, 561113, Guiyang, Guizhou, P.R. China.
| |
Collapse
|
4
|
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
|
5
|
Beito M, Taegtmeyer H. Metabolic cycles: A unifying concept for energy transfer in the heart. J Mol Cell Cardiol 2024; 195:103-109. [PMID: 39154711 DOI: 10.1016/j.yjmcc.2024.08.002] [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: 06/11/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
It is still debated whether changes in metabolic flux are cause or consequence of contractile dysfunction in non-ischemic heart disease. We have previously proposed a model of cardiac metabolism grounded in a series of six moiety-conserved, interconnected cycles. In view of a recent interest to augment oxygen availability in heart failure through iron supplementation, we integrated this intervention in terms of moiety conservation. Examining published work from both human and murine models, we argue this strategy restores a mitochondrial cycle of energy transfer by enhancing mitochondrial pyruvate carrier (MPC) expression and providing pyruvate as a substrate for carboxylation and anaplerosis. Metabolomic data from failing heart muscle reveal elevated pyruvate levels with a concomitant decrease in the levels of Krebs cycle intermediates. Additionally, MPC is downregulated in the same failing hearts, as well as under hypoxic conditions. MPC expression increases upon mechanical unloading in the failing human heart, as does contractile function. We note that MPC deficiency also alters expression of enzymes involved in pyruvate carboxylation and decarboxylation, increases intermediates of biosynthetic pathways, and eventually leads to cardiac hypertrophy and dilated cardiomyopathy. Collectively, we propose that an unbroken chain of moiety-conserved cycles facilitates energy transfer in the heart. We refer to the transport and subsequent carboxylation of pyruvate in the mitochondrial matrix as an example and a proposed target for metabolic support to reverse impaired contractile function.
Collapse
Affiliation(s)
- Mitchell Beito
- McGovern Medical School - The University of Texas Health Science Center at Houston, United States of America
| | - Heinrich Taegtmeyer
- McGovern Medical School - The University of Texas Health Science Center at Houston Department of Internal Medicine, Division of Cardiology, 6431 Fannin St. Houston, TX 77030, United States of America.
| |
Collapse
|
6
|
Wang H, Cui W, Yue S, Zhu X, Li X, He L, Zhang M, Yang Y, Wei M, Wu H, Wang S. Malic enzymes in cancer: Regulatory mechanisms, functions, and therapeutic implications. Redox Biol 2024; 75:103273. [PMID: 39142180 PMCID: PMC11367648 DOI: 10.1016/j.redox.2024.103273] [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: 03/28/2024] [Revised: 05/21/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Malic enzymes (MEs) are metabolic enzymes that catalyze the oxidation of malate to pyruvate and NAD(P)H. While researchers have well established the physiological metabolic roles of MEs in organisms, recent research has revealed a link between MEs and carcinogenesis. This review collates evidence of the molecular mechanisms by which MEs promote cancer occurrence, including transcriptional regulation, post-transcriptional regulation, post-translational protein modifications, and protein-protein interactions. Additionally, we highlight the roles of MEs in reprogramming energy metabolism, suppressing senescence, and modulating the tumor immune microenvironment. We also discuss the involvement of these enzymes in mediating tumor resistance and how the development of novel small-molecule inhibitors targeting MEs might be a good therapeutic approach. Insights through this review are expected to provide a comprehensive understanding of the intricate relationship between MEs and cancer, while facilitating future research on the potential therapeutic applications of targeting MEs in cancer management.
Collapse
Affiliation(s)
- Huan Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, PR China.
| | - Wanlin Cui
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, PR China.
| | - Song Yue
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, Liaoning Province, PR China.
| | - Xianglong Zhu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, PR China
| | - Xiaoyan Li
- Department of Pathology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, PR China
| | - Lian He
- Department of Pathology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, PR China
| | - Mingrong Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, PR China
| | - Yan Yang
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, No.4, Chongshan Road, Huanggu District, Shenyang, Liaoning Province, PR China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, PR China; Shenyang Kangwei Medical Laboratory Analysis Co. LTD, Shenyang City, Liaoning Province, PR China.
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, PR China.
| | - Shuo Wang
- Department of Gynecology Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, PR China.
| |
Collapse
|
7
|
Huang Y, Yang Y, Chen X, Zeng S, Chen Y, Wang H, Lv X, Hu X, Teng L. Downregulation of malic enzyme 3 facilitates progression of gastric carcinoma via regulating intracellular oxidative stress and hypoxia-inducible factor-1α stabilization. Cell Mol Life Sci 2024; 81:375. [PMID: 39212717 PMCID: PMC11364750 DOI: 10.1007/s00018-024-05388-9] [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: 01/31/2024] [Revised: 07/04/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most malignant cancers worldwide. Metabolism disorder is a critical characteristic of malignant tumors related to tumor progression and metastasis. However, the expression and molecular mechanism of malic enzyme 3 (ME3) in GC are rarely reported. In this study, we aim to investigate the molecular mechanism of ME3 in the development of GC and to explore its potential value as a prognostic and therapeutic target in GC. METHOD ME3 mRNA and protein expression were evaluated in patients with GC using RT-qPCR, WB, and immunohistochemistry, as well as their correlation with clinicopathological indicators. The effect of ME3 on proliferation and metastasis was evaluated using Cell Counting Kit-8 (CCK-8), 5-ethynyl-20-deoxyuridine (EdU) assay, transwell assay, wound healing assay, and subcutaneous injection or tail vein injection of tumor cells in mice model. The effects of ME3 knockdown on the level of metabolites and hypoxia-inducible factor-1α (HIF-1α) protein were determined in GC cells. Oxidative phosphorylation was measured to evaluate adenosine triphosphate (ATP) production. RESULTS ME3 was downregulated in human GC tissues (P < 0.001). The decreased ME3 mRNA expression was associated with younger age (P = 0.02), pathological staging (P = 0.049), and lymph node metastasis (P = 0.001), while low ME3 expression was associated with tumor size (P = 0.048), tumor invasion depth (P < 0.001), lymph node metastasis (P = 0.018), TNM staging (P < 0.001), and poor prognosis (OS, P = 0.0206; PFS P = 0.0453). ME3 knockdown promoted GC cell malignancy phenotypes. Moreover, α-ketoglutarate (α-KG) and NADPH/NADP+ ratios were reduced while malate was increased in the ME3 knockdown group under normoxia. When cells were incubated under hypoxia, the NADPH/NADP+ ratio and α-KG decreased while intracellular reactive oxygen species (ROS) increased significantly. The ME3 knockdown group exhibited an increase in ATP production and while ME3 overexpression group exhibited oppositely. We discovered that ME3 and HIF-1α expression were negatively correlated in GC cells and tissues, and proposed the hypothesis: downregulation of ME3 promotes GC progression via regulating intracellular oxidative stress and HIF-1α. CONCLUSION We provide evidence that ME3 downregulation is associated with poor prognosis in GC patients and propose a hypothesis for the ME3 regulatory mechanism in GC progression. The present study is of great scientific significance and clinical value for exploring the prognostic and therapeutic targets of GC, evaluating and improving the clinical efficacy of patients, reducing recurrence and metastasis, and improving the prognosis and quality of life of patients.
Collapse
Affiliation(s)
- Yingying Huang
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Gynecology, Guangzhou First People's Hospital, Guangzhou, China
| | - Yan Yang
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiangliu Chen
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Siying Zeng
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiran Chen
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haiyong Wang
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiadong Lv
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xun Hu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lisong Teng
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
Alektiar JM, Shan M, Radyk MD, Zhang L, Halbrook CJ, Lin L, Espinoza C, Mier IF, Lavoie BL, Salvatore L, Pasca di Magliano M, Cantley LC, Mueller JL, Lyssiotis CA. Malic enzyme 1 knockout has no deleterious phenotype and is favored in the male germline under standard laboratory conditions. PLoS One 2024; 19:e0303577. [PMID: 38843233 PMCID: PMC11156412 DOI: 10.1371/journal.pone.0303577] [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: 07/06/2023] [Accepted: 04/26/2024] [Indexed: 06/09/2024] Open
Abstract
Malic Enzyme 1 (ME1) plays an integral role in fatty acid synthesis and cellular energetics through its production of NADPH and pyruvate. As such, it has been identified as a gene of interest in obesity, type 2 diabetes, and an array of epithelial cancers, with most work being performed in vitro. The current standard model for ME1 loss in vivo is the spontaneous Mod-1 null allele, which produces a canonically inactive form of ME1. Herein, we describe two new genetically engineered mouse models exhibiting ME1 loss at dynamic timepoints. Using murine embryonic stem cells and Flp/FRT and Cre/loxP class switch recombination, we established a germline Me1 knockout model (Me1 KO) and an inducible conditional knockout model (Me1 cKO), activated upon tamoxifen treatment in adulthood. Collectively, neither the Me1 KO nor Me1 cKO models exhibited deleterious phenotype under standard laboratory conditions. Knockout of ME1 was validated by immunohistochemistry and genotype confirmed by PCR. Transmission patterns favor Me1 loss in Me1 KO mice when maternally transmitted to male progeny. Hematological examination of these models through complete blood count and serum chemistry panels revealed no discrepancy with their wild-type counterparts. Orthotopic pancreatic tumors in Me1 cKO mice grow similarly to Me1 expressing mice. Similarly, no behavioral phenotype was observed in Me1 cKO mice when aged for 52 weeks. Histological analysis of several tissues revealed no pathological phenotype. These models provide a more modern approach to ME1 knockout in vivo while opening the door for further study into the role of ME1 loss under more biologically relevant, stressful conditions.
Collapse
Affiliation(s)
- Jonathan M. Alektiar
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Mengrou Shan
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Megan D. Radyk
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Li Zhang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Christopher J. Halbrook
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lin Lin
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Carlos Espinoza
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ivan F. Mier
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Brooke L. Lavoie
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lucie Salvatore
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lewis C. Cantley
- Department of Cancer Biology, Dana Farber Cancer Center, Boston, Massachusetts, United States of America
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jacob L. Mueller
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Costas A. Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
9
|
Oku Y, Matsuda T. Substrate Promiscuity of Thermoplasma acidophilum Malic Enzyme for CO 2 Fixation Reaction. JACS AU 2024; 4:1758-1762. [PMID: 38818066 PMCID: PMC11134350 DOI: 10.1021/jacsau.4c00290] [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: 04/01/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 06/01/2024]
Abstract
CO2 fixation technology has gained attention as a method to effectively utilize the abundant CO2 in the atmosphere by converting it into useful chemicals. However, since CO2 is a highly stable molecule, many of the currently developed methods for chemical CO2 fixation require harsh conditions and reactive reagents. The establishment of efficient and sustainable processes is eagerly awaited. In this study, we investigated a biocatalytic process and achieved a carboxylation reaction under mild conditions (37 °C, 0.1 MPa CO2) using a biocatalyst, Thermoplasma acidophilum NADP+-malic enzyme (TaME), and gaseous CO2 by coupling enzymatic coenzyme regeneration. We also demonstrated for the first time that the carboxylation reaction by ME proceeds not only with pyruvate, a natural substrate, but also with 2-ketoglutarate.
Collapse
Affiliation(s)
- Yuri Oku
- Department of Life Science
and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, JAPAN
| | - Tomoko Matsuda
- Department of Life Science
and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, JAPAN
| |
Collapse
|
10
|
Lee S. Cardiovascular Disease and miRNAs: Possible Oxidative Stress-Regulating Roles of miRNAs. Antioxidants (Basel) 2024; 13:656. [PMID: 38929095 PMCID: PMC11200533 DOI: 10.3390/antiox13060656] [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: 04/19/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
MicroRNAs (miRNAs) have been highlighted as key players in numerous diseases, and accumulating evidence indicates that pathological expressions of miRNAs contribute to both the development and progression of cardiovascular diseases (CVD), as well. Another important factor affecting the development and progression of CVD is reactive oxygen species (ROS), as well as the oxidative stress they may impose on the cells. Considering miRNAs are involved in virtually every biological process, it is not unreasonable to assume that miRNAs also play critical roles in the regulation of oxidative stress. This narrative review aims to provide mechanistic insights on possible oxidative stress-regulating roles of miRNAs in cardiovascular diseases based on differentially expressed miRNAs reported in various cardiovascular diseases and their empirically validated targets that have been implicated in the regulation of oxidative stress.
Collapse
Affiliation(s)
- Seahyoung Lee
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| |
Collapse
|
11
|
Li X, Zhang FG, Ma JA, Liu Y. Computational insights into the binding modes, keto-enol tautomerization and stereo-electronically controlled decarboxylation of oxaloacetate in the active site of macrophomate synthase. Phys Chem Chem Phys 2024; 26:12331-12344. [PMID: 38598177 DOI: 10.1039/d4cp00716f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Oxaloacetic acid (OAA) is a β-ketocarboxylic acid, which plays an important role as an intermediate in some metabolic pathways, including the tricarboxylic acid cycle, gluconeogenesis and fatty acid biosynthesis. Animal studies have indicated that supplementing oxaloacetic acid shows an increase of lifespan and other substantial health benefits including mitochondrial DNA protection, and protection of retinal, neural and pancreatic tissues. Most of the chemical transformations of OAA in the metabolic pathways have been extensively studied; however, the understanding of decarboxylation of OAA at the atomic level is relatively lacking. Here, we carried out MD simulations and combined quantum mechanical/molecular mechanical (QM/MM) calculations as an example to systematically elucidate the binding modes, keto-enol tautomerization and decarboxylation of OAA in the active site of macrophomate synthase (MPS), which is a Mg(II)-dependent bifunctional enzyme that catalyzes both the decarboxylation of OAA and [4+2] cycloaddition of 2-pyrone with the decarboxylated intermediate of OAA (pyruvate enolate). On the basis of our calculations, it was found that the Mg2+-coordinated oxaloacetate may exist in enol forms and keto forms. The four keto forms can be transformed into each other by simply rotating the C2-C3 single bond, nevertheless, the keto-enol tautomerization strictly requires the assistance of pocket water molecules. In addition, the decarboxylation is stereo-electronically controlled, i.e., it is the relative orientation of the terminal carboxyl anion that determines the rate of decarboxylation. As such, the chemistry of oxaloacetate in the active site of MPS is complex. On one hand, the most stable binding mode (K-I) may undergo enol-keto tautomerization to isomerize to the enol form, which may further react with the second substrate; on the other hand, K-I may isomerize to another binding mode K-II to proceed decarboxylation to generate pyruvate enolate and CO2. Starting from K-I, the enol-keto tautomerization corresponds to a barrier of 16.2 kcal mol-1, whereas the decarboxylation is associated with an overall barrier of 19.7 kcal mol-1. These findings may provide useful information for understanding the chemistry of OAA and the catalysis of related enzymes, and they are basically in agreement with the available experimental kinetic data.
Collapse
Affiliation(s)
- Xinyi Li
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| | - Fa-Guang Zhang
- Department of Chemistry, Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.
| | - Jun-An Ma
- Department of Chemistry, Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.
| | - Yongjun Liu
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| |
Collapse
|
12
|
Luo Y, Qi X, Zhang Z, Zhang J, Li B, Shu T, Li X, Hu H, Li J, Tang Q, Zhou Y, Wang M, Fan T, Guo W, Liu Y, Zhang J, Pang J, Yang P, Gao R, Chen W, Yan C, Xing Y, Du W, Wang J, Wang C. Inactivation of Malic Enzyme 1 in Endothelial Cells Alleviates Pulmonary Hypertension. Circulation 2024; 149:1354-1371. [PMID: 38314588 DOI: 10.1161/circulationaha.123.067579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive cardiopulmonary disease with a high mortality rate. Although growing evidence has revealed the importance of dysregulated energetic metabolism in the pathogenesis of PH, the underlying cellular and molecular mechanisms are not fully understood. In this study, we focused on ME1 (malic enzyme 1), a key enzyme linking glycolysis to the tricarboxylic acid cycle. We aimed to determine the role and mechanistic action of ME1 in PH. METHODS Global and endothelial-specific ME1 knockout mice were used to investigate the role of ME1 in hypoxia- and SU5416/hypoxia (SuHx)-induced PH. Small hairpin RNA and ME1 enzymatic inhibitor (ME1*) were used to study the mechanism of ME1 in pulmonary artery endothelial cells. Downstream key metabolic pathways and mediators of ME1 were identified by metabolomics analysis in vivo and ME1-mediated energetic alterations were examined by Seahorse metabolic analysis in vitro. The pharmacological effect of ME1* on PH treatment was evaluated in PH animal models induced by SuHx. RESULTS We found that ME1 protein level and enzymatic activity were highly elevated in lung tissues of patients and mice with PH, primarily in vascular endothelial cells. Global knockout of ME1 protected mice from developing hypoxia- or SuHx-induced PH. Endothelial-specific ME1 deletion similarly attenuated pulmonary vascular remodeling and PH development in mice, suggesting a critical role of endothelial ME1 in PH. Mechanistic studies revealed that ME1 inhibition promoted downstream adenosine production and activated A2AR-mediated adenosine signaling, which leads to an increase in nitric oxide generation and a decrease in proinflammatory molecule expression in endothelial cells. ME1 inhibition activated adenosine production in an ATP-dependent manner through regulating malate-aspartate NADH (nicotinamide adenine dinucleotide plus hydrogen) shuttle and thereby balancing oxidative phosphorylation and glycolysis. Pharmacological inactivation of ME1 attenuated the progression of PH in both preventive and therapeutic settings by promoting adenosine production in vivo. CONCLUSIONS Our findings indicate that ME1 upregulation in endothelial cells plays a causative role in PH development by negatively regulating adenosine production and subsequently dysregulating endothelial functions. Our findings also suggest that ME1 may represent as a novel pharmacological target for upregulating protective adenosine signaling in PH therapy.
Collapse
Affiliation(s)
- Ya Luo
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
- Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.L.)
| | - Xianmei Qi
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Zhenxi Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases (Z.Z., W.D.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jiawei Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Bolun Li
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Ting Shu
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Xiaona Li
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Huiyuan Hu
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jinqiu Li
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Qihao Tang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Yitian Zhou
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Mingyao Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China (M.W., C.W.)
| | - Tianfei Fan
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Wenjun Guo
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Ying Liu
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jin Zhang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China (J.Z.)
| | - Junling Pang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Ran Gao
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Wenhui Chen
- Department of Lung Transplantation, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China (W.C.)
| | - Chen Yan
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (C.Y.)
| | - Yanjiang Xing
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Wenjing Du
- State Key Laboratory of Common Mechanism Research for Major Diseases (Z.Z., W.D.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jing Wang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Chen Wang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China (M.W., C.W.)
- Chinese Academy of Engineering, Beijing, China (C.W.)
| |
Collapse
|
13
|
Scott KM, Payne RR, Gahramanova A. Widespread dissolved inorganic carbon-modifying toolkits in genomes of autotrophic Bacteria and Archaea and how they are likely to bridge supply from the environment to demand by autotrophic pathways. Appl Environ Microbiol 2024; 90:e0155723. [PMID: 38299815 PMCID: PMC10880623 DOI: 10.1128/aem.01557-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024] Open
Abstract
Using dissolved inorganic carbon (DIC) as a major carbon source, as autotrophs do, is complicated by the bedeviling nature of this substance. Autotrophs using the Calvin-Benson-Bassham cycle (CBB) are known to make use of a toolkit comprised of DIC transporters and carbonic anhydrase enzymes (CA) to facilitate DIC fixation. This minireview provides a brief overview of the current understanding of how toolkit function facilitates DIC fixation in Cyanobacteria and some Proteobacteria using the CBB and continues with a survey of the DIC toolkit gene presence in organisms using different versions of the CBB and other autotrophic pathways (reductive citric acid cycle, Wood-Ljungdahl pathway, hydroxypropionate bicycle, hydroxypropionate-hydroxybutyrate cycle, and dicarboxylate-hydroxybutyrate cycle). The potential function of toolkit gene products in these organisms is discussed in terms of CO2 and HCO3- supply from the environment and demand by the autotrophic pathway. The presence of DIC toolkit genes in autotrophic organisms beyond those using the CBB suggests the relevance of DIC metabolism to these organisms and provides a basis for better engineering of these organisms for industrial and agricultural purposes.
Collapse
Affiliation(s)
- Kathleen M. Scott
- Integrative Biology Department, University of South Florida, Tampa, Florida, USA
| | - Ren R. Payne
- Integrative Biology Department, University of South Florida, Tampa, Florida, USA
| | - Arin Gahramanova
- Integrative Biology Department, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
14
|
Chen T, Xie S, Cheng J, Zhao Q, Wu H, Jiang P, Du W. AKT1 phosphorylation of cytoplasmic ME2 induces a metabolic switch to glycolysis for tumorigenesis. Nat Commun 2024; 15:686. [PMID: 38263319 PMCID: PMC10805786 DOI: 10.1038/s41467-024-44772-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 01/04/2024] [Indexed: 01/25/2024] Open
Abstract
Many types of tumors feature aerobic glycolysis for meeting their increased energetic and biosynthetic demands. However, it remains still unclear how this glycolytic phenomenon is achieved and coordinated with other metabolic pathways in tumor cells in response to growth stimuli. Here we report that activation of AKT1 induces a metabolic switch to glycolysis from the mitochondrial metabolism via phosphorylation of cytoplasmic malic enzyme 2 (ME2), named ME2fl (fl means full length), favoring an enhanced glycolytic phenotype. Mechanistically, in the cytoplasm, AKT1 phosphorylates ME2fl at serine 9 in the mitochondrial localization signal peptide at the N-terminus, preventing its mitochondrial translocation. Unlike mitochondrial ME2, which accounts for adjusting the tricarboxylic acid (TCA) cycle, ME2fl functions as a scaffold that brings together the key glycolytic enzymes phosphofructokinase (PFKL), glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and pyruvate kinase M2 (PKM2), as well as Lactate dehydrogenase A (LDHA), to promote glycolysis in the cytosol. Thus, through phosphorylation of ME2fl, AKT1 enhances the glycolytic capacity of tumor cells in vitro and in vivo, revealing an unexpected role for subcellular translocation switching of ME2 mediated by AKT1 in the metabolic adaptation of tumor cells to growth stimuli.
Collapse
Affiliation(s)
- Taiqi Chen
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), South Medical University, Guangzhou, 510080, China
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Siyi Xie
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Jie Cheng
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Qiao Zhao
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hong Wu
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
- School of Life Sciences, Peking University, Beijing, 100084, China.
| | - Peng Jiang
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), South Medical University, Guangzhou, 510080, China.
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
| | - Wenjing Du
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
15
|
Moreira R, Martins AD, Ferreira R, Alves MG, Pereira MDL, Oliveira PF. Impact of Chromium Picolinate on Leydig Cell Steroidogenesis and Antioxidant Balance Using an In Vitro Insulin Resistance Model. Antioxidants (Basel) 2023; 13:40. [PMID: 38247463 PMCID: PMC10812815 DOI: 10.3390/antiox13010040] [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: 11/15/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Leydig cells (LCs) play a pivotal role in male fertility, producing testosterone. Chromium (III) picolinate (CrPic3), a contentious supplement with antidiabetic and antioxidant properties, raises concerns regarding male fertility. Using a rodent LC line, we investigated the cytotoxicity of increasing CrPic3 doses. An insulin resistance (IR) model was established using palmitate (PA), and LCs were further exposed to CrPic3 to assess its antioxidant/antidiabetic activities. An exometabolome analysis was performed using 1H-NMR. Mitochondrial function and oxidative stress were evaluated via immunoblot. Steroidogenesis was assessed by quantifying androstenedione through ELISA. Our results uncover the toxic effects of CrPic3 on LCs even at low doses under IR conditions. Furthermore, even under these IR conditions, CrPic3 fails to enhance glucose consumption but restores the expression of mitochondrial complexes CII and CIII, alleviating oxidative stress in LCs. While baseline androgen production remained unaffected, CrPic3 promoted androstenedione production in LCs in the presence of PA, suggesting that it promotes cholesterol conversion into androgenic intermediates in this context. This study highlights the need for caution with CrPic3 even at lower doses. It provides valuable insights into the intricate factors influencing LCs metabolism and antioxidant defenses, shedding light on potential benefits and risks of CrPic3, particularly in IR conditions.
Collapse
Affiliation(s)
- Rúben Moreira
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (R.M.); (A.D.M.); (R.F.)
- LAQV-REQUIMTE, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana D. Martins
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (R.M.); (A.D.M.); (R.F.)
- LAQV-REQUIMTE, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rita Ferreira
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (R.M.); (A.D.M.); (R.F.)
- LAQV-REQUIMTE, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Marco G. Alves
- iBiMED-Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal;
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Pedro F. Oliveira
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (R.M.); (A.D.M.); (R.F.)
- LAQV-REQUIMTE, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
16
|
Afzal AR, Jeon J, Jung CH. Fumarase activity in NAD-dependent malic enzyme, MaeA, from Escherichia coli. Biochem Biophys Res Commun 2023; 678:144-147. [PMID: 37634412 DOI: 10.1016/j.bbrc.2023.08.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/21/2023] [Indexed: 08/29/2023]
Abstract
NAD-dependent malic enzymes catalyze NAD reduction to NADH while converting malate to pyruvate and CO2. In this study, NAD was reduced to NADH by MaeA, NAD-dependent malic enzyme from Escherichia coli, when fumarate was used as substrate. This suggested that MaeA catalyzed the conversion of fumarate to malate and then malate to pyruvate. The K0.5 value for fumarate was determined as 13 mM, different from previously characterized fumarases in Escherichia coli. Fumarate inhibited the malic enzyme activity of MaeA where NAD reduction to NADH was examined in the presence of malate as substrate. Human ME2, an NAD-dependent malic enzyme, also converted NAD to NADH in the presence of fumarate, suggesting that the duplex activity as fumarase and malic enzyme might be conserved in various NAD-dependent malic enzymes. MaeB, NADP-dependent malic enzyme from Escherichia coli, did not reduce NADP to NADPH in the presence of fumarate, suggesting the fumarase activities of MaeA and ME2 were specific.
Collapse
Affiliation(s)
- Aqeel Rana Afzal
- Department of Medical Science, Chonam National University, Gwangju, 61186, South Korea
| | - Jinyoung Jeon
- Department of Medical Science, Chonam National University, Gwangju, 61186, South Korea
| | - Che-Hun Jung
- Department of Medical Science, Chonam National University, Gwangju, 61186, South Korea; Department of Chemistry, Chonnam National University, Gwangju, 61186, South Korea.
| |
Collapse
|
17
|
Wu B, Qi F, Liang Y. Fuels for ROS signaling in plant immunity. TRENDS IN PLANT SCIENCE 2023; 28:1124-1131. [PMID: 37188557 DOI: 10.1016/j.tplants.2023.04.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023]
Abstract
Reactive oxygen species (ROS) signaling has an important role in plant innate immune responses and is primarily mediated by NADPH oxidase, also known as respiratory burst oxidase homologs (RBOHs) in plants. NADPH serves as a fuel for RBOHs and limits the rate or amount of ROS production. Molecular regulation of RBOHs has been extensively studied; however, the source of NADPH for RBOHs has received little attention. Here, we review ROS signaling and the regulation of RBOHs in the plant immune system with a focus on NADPH regulation to achieve ROS homeostasis. We propose an idea to regulate the levels of NADPH as part of a new strategy to control ROS signaling and the corresponding downstream defense responses.
Collapse
Affiliation(s)
- Binyan Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Fan Qi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Yan Liang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
18
|
Yang C, Fan H, Ge L, Ma Q, Jiang M, Wen H. Comparative analysis of quantitative phosphoproteomics between two tilapias ( Oreochromis niloticus and Oreochromis aureus) under low-temperature stress. PeerJ 2023; 11:e15599. [PMID: 37456864 PMCID: PMC10340112 DOI: 10.7717/peerj.15599] [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: 02/02/2023] [Accepted: 05/30/2023] [Indexed: 07/18/2023] Open
Abstract
As an important farmed fish, tilapia has poor tolerance to low-temperatures. At the same time, different tilapia strains have apparent differences in low-temperature tolerance. In this study, using the iTRAQ method, the phosphorylated proteomics of two tilapia strains (Oreochromis niloticus and Oreochromis aureus) with different tolerances to low-temperature stress were quantitatively and comparatively analyzed, to clarify the physiological mechanism of tilapia's response to low-temperature stress. Through the GO and IPR analyses of differentially phosphorylated proteins, a number of similarities in physiological activities and regulatory effects were found between the two tilapias in response to low-temperature stress. Many differentially phosphorylated proteins are mainly involved in lipid metabolism, cell proliferation and apoptosis. However, the difference in endurance of low temperature of these two tilapias might be related to the differences in categories, expression and modification level of genetic products which were involved in the aforementioned physiological processes. And meanwhile, the enrichment results of KEGG showed the changes of multiple immune-related and growth-related phosphorylated proteins in the cytokine-cytokine receptor interaction pathway in O. aureus are more prominent. Furthermore, the significantly enriched pathway of carbohydrate digestion and absorption in O. niloticus may indicate that low-temperature stress exerts a more severe impact on energy metabolism. The relative results would help elucidating the molecular mechanism by which tilapia responds to low-temperature stress, and developing culture of tilapia species.
Collapse
Affiliation(s)
- Changgeng Yang
- Life Science & Technology School, Lingnan Normal University, Zhanjiang, China
| | - Hua Fan
- Life Science & Technology School, Lingnan Normal University, Zhanjiang, China
| | - Liya Ge
- Life Science & Technology School, Lingnan Normal University, Zhanjiang, China
| | - Qian Ma
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Ming Jiang
- Fish Nutrition and Feed Division, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
| | - Hua Wen
- Fish Nutrition and Feed Division, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
| |
Collapse
|
19
|
Li H, Zhang L, Wu B, Li Y, Wang H, Teng H, Wei D, Yuan Z, Yuan Z. Physiological and proteomic analyses reveal the important role of arbuscular mycorrhizal fungi on enhancing photosynthesis in wheat under cadmium stress. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 261:115105. [PMID: 37285679 DOI: 10.1016/j.ecoenv.2023.115105] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/20/2023] [Accepted: 06/02/2023] [Indexed: 06/09/2023]
Abstract
Arbuscular mycorrhizal fungi (AMF) are important in the phytoremediation of cadmium (Cd). Improving photosynthesis under Cd stress helps to increase crop yields. However, the molecular regulatory mechanisms of AMF on photosynthetic processes in wheat (Triticum aestivum) under Cd stress remain unclear. This study utilized physiological and proteomic analyses to reveal the key processes and related genes of AMF that regulate photosynthesis under Cd stress. The results showed that AMF promoted the accumulation of Cd in the roots of wheat but significantly reduced the content of Cd in the shoots and grains. The photosynthetic rates, stomatal conductance, transpiration rates, chlorophyll content, and accumulation of carbohydrates under Cd stress were increased by AMF symbiosis. Proteomic analysis showed that AMF significantly induced the expression of two enzymes involved in the chlorophyll biosynthetic pathway (coproporphyrinogen oxidase and Mg-protoporphyrin IX chelatase), improved the expression of two proteins related to CO2 assimilation (ribulose-1,5-bisphosphate carboxylase and malic enzyme), and increased the expression of S-adenosylmethionine synthase, which positively regulates abiotic stress. Therefore, AMF may regulate photosynthesis under Cd stress by promoting chlorophyll biosynthesis, carbon assimilation, and S-adenosylmethionine metabolism.
Collapse
Affiliation(s)
- Hua Li
- College of Life Science, Henan Agricultural University, Zhengzhou, China
| | - Lele Zhang
- College of Life Science, Henan Agricultural University, Zhengzhou, China
| | - Baocun Wu
- Henan Province Rock & Mineral Testing Centre, Zhengzhou, China; Laboratory of Precious Metal Analysis and Exploration Technology, Ministry of Natural Resources, Zhengzhou, China
| | - Yang Li
- College of Life Science, Henan Agricultural University, Zhengzhou, China
| | - Huijuan Wang
- College of Life Science, Henan Agricultural University, Zhengzhou, China
| | - Huixin Teng
- College of Life Science, Henan Agricultural University, Zhengzhou, China
| | - Dongwei Wei
- College of Life Science, Henan Agricultural University, Zhengzhou, China.
| | - Zhiliang Yuan
- College of Life Science, Henan Agricultural University, Zhengzhou, China.
| | - Zuli Yuan
- College of Life Science, Henan Agricultural University, Zhengzhou, China.
| |
Collapse
|
20
|
Hsieh JY, Chen KC, Wang CH, Liu GY, Ye JA, Chou YT, Lin YC, Lyu CJ, Chang RY, Liu YL, Li YH, Lee MR, Ho MC, Hung HC. Suppression of the human malic enzyme 2 modifies energy metabolism and inhibits cellular respiration. Commun Biol 2023; 6:548. [PMID: 37217557 DOI: 10.1038/s42003-023-04930-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 05/12/2023] [Indexed: 05/24/2023] Open
Abstract
Human mitochondrial NAD(P)+-dependent malic enzyme (ME2) is well-known for its role in cell metabolism, which may be involved in cancer or epilepsy. We present potent ME2 inhibitors based on cyro-EM structures that target ME2 enzyme activity. Two structures of ME2-inhibitor complexes demonstrate that 5,5'-Methylenedisalicylic acid (MDSA) and embonic acid (EA) bind allosterically to ME2's fumarate-binding site. Mutagenesis studies demonstrate that Asn35 and the Gln64-Tyr562 network are required for both inhibitors' binding. ME2 overexpression increases pyruvate and NADH production while decreasing the cell's NAD+/NADH ratio; however, ME2 knockdown has the opposite effect. MDSA and EA inhibit pyruvate synthesis and thus increase the NAD+/NADH ratio, implying that these two inhibitors interfere with metabolic changes by inhibiting cellular ME2 activity. ME2 silence or inhibiting ME2 activity with MDSA or EA decreases cellular respiration and ATP synthesis. Our findings suggest that ME2 is crucial for mitochondrial pyruvate and energy metabolism, as well as cellular respiration, and that ME2 inhibitors could be useful in the treatment of cancer or other diseases that involve these processes.
Collapse
Affiliation(s)
- Ju-Yi Hsieh
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan ROC
| | - Kun-Chi Chen
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan ROC
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, 402, Taiwan ROC
| | - Chun-Hsiung Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115, Taiwan ROC
| | - Guang-Yaw Liu
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung, 402, Taiwan ROC
| | - Jie-An Ye
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan ROC
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung, 402, Taiwan ROC
| | - Yu-Tung Chou
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan ROC
| | - Yi-Chun Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan ROC
| | - Cheng-Jhe Lyu
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan ROC
| | - Rui-Ying Chang
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan ROC
| | - Yi-Liang Liu
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan ROC
| | - Yen-Hsien Li
- Instrument Center, Office of Research and Development, National Chung Hsing University, Taichung, 40227, Taiwan ROC
- Department of Chemistry, National Chung Hsing University, Taichung, 402, Taiwan ROC
| | - Mau-Rong Lee
- Department of Chemistry, National Chung Hsing University, Taichung, 402, Taiwan ROC
| | - Meng-Chiao Ho
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115, Taiwan ROC.
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 106, Taiwan ROC.
| | - Hui-Chih Hung
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan ROC.
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, 402, Taiwan ROC.
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, 402, Taiwan ROC.
- Advanced Plant and Food Crop Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan ROC.
| |
Collapse
|
21
|
Xiong L, Pei J, Bao P, Wang X, Guo S, Cao M, Kang Y, Yan P, Guo X. The Effect of the Feeding System on Fat Deposition in Yak Subcutaneous Fat. Int J Mol Sci 2023; 24:ijms24087381. [PMID: 37108542 PMCID: PMC10138426 DOI: 10.3390/ijms24087381] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Fat deposition is very important to the growth and reproduction of yaks. In this study, the effect of the feeding system on fat deposition in yaks was explored by transcriptomics and lipidomics. The thickness of the subcutaneous fat in yaks under stall (SF) and graze feeding (GF) was evaluated. The transcriptomes and lipidomes of the subcutaneous fat in yaks under different feeding systems were detected by RNA-sequencing (RNA-Seq) and non-targeted lipidomics based on ultrahigh-phase liquid chromatography tandem mass spectrometry (UHPLC-MS), respectively. The differences in lipid metabolism were explored, and the function of differentially expressed genes (DEGs) was evaluated by gene ontology (GO) and Kyoto encyclopedia of genes and genome (KEGG) analysis. Compared with GF yaks, SF yaks possessed stronger fat deposition capacity. The abundance of 12 triglycerides (TGs), 3 phosphatidylethanolamines (PEs), 3 diglycerides (DGs), 2 sphingomyelins (SMs) and 1 phosphatidylcholine (PC) in the subcutaneous fat of SF and GF yaks was significantly different. Under the mediation of the cGMP-PKG signaling pathway, the blood volume of SF and GF yaks may be different, which resulted in the different concentrations of precursors for fat deposition, including non-esterified fatty acid (NEFA), glucose (GLU), TG and cholesterol (CH). The metabolism of C16:0, C16:1, C17:0, C18:0, C18:1, C18:2 and C18:3 in yak subcutaneous fat was mainly realized under the regulation of the INSIG1, ACACA, FASN, ELOVL6 and SCD genes, and TG synthesis was regulated by the AGPAT2 and DGAT2 genes. This study will provide a theoretical basis for yak genetic breeding and healthy feeding.
Collapse
Affiliation(s)
- Lin Xiong
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China
| | - Jie Pei
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China
| | - Pengjia Bao
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China
| | - Xingdong Wang
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China
| | - Shaoke Guo
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China
| | - Mengli Cao
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China
| | - Yandong Kang
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China
| | - Ping Yan
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China
| | - Xian Guo
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China
| |
Collapse
|
22
|
Fan S, Chen J, Yang R. Candidate Genes for Salt Tolerance in Forage Sorghum under Saline Conditions from Germination to Harvest Maturity. Genes (Basel) 2023; 14:genes14020293. [PMID: 36833220 PMCID: PMC9956952 DOI: 10.3390/genes14020293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/23/2022] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
To address the plant adaptability of sorghum (Sorghum bicolor) in salinity, the research focus should shift from only selecting tolerant varieties to understanding the precise whole-plant genetic coping mechanisms with long-term influence on various phenotypes of interest to expanding salinity, improving water use, and ensuring nutrient use efficiency. In this review, we discovered that multiple genes may play pleiotropic regulatory roles in sorghum germination, growth, and development, salt stress response, forage value, and the web of signaling networks. The conserved domain and gene family analysis reveals a remarkable functional overlap among members of the bHLH (basic helix loop helix), WRKY (WRKY DNA-binding domain), and NAC (NAM, ATAF1/2, and CUC2) superfamilies. Shoot water and carbon partitioning, for example, are dominated by genes from the aquaporins and SWEET families, respectively. The gibberellin (GA) family of genes is prevalent during pre-saline exposure seed dormancy breaking and early embryo development at post-saline exposure. To improve the precision of the conventional method of determining silage harvest maturity time, we propose three phenotypes and their underlying genetic mechanisms: (i) the precise timing of transcriptional repression of cytokinin biosynthesis (IPT) and stay green (stg1 and stg2) genes; (ii) the transcriptional upregulation of the SbY1 gene and (iii) the transcriptional upregulation of the HSP90-6 gene responsible for grain filling with nutritive biochemicals. This work presents a potential resource for sorghum salt tolerance and genetic studies for forage and breeding.
Collapse
|
23
|
Grell TA, Mason M, Thompson AA, Gómez-Tamayo JC, Riley D, Wagner MV, Steele R, Ortiz-Meoz RF, Wadia J, Shaffer PL, Tresadern G, Sharma S, Yu X. Integrative structural and functional analysis of human malic enzyme 3: A potential therapeutic target for pancreatic cancer. Heliyon 2022; 8:e12392. [PMID: 36590518 PMCID: PMC9801130 DOI: 10.1016/j.heliyon.2022.e12392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Malic enzymes (ME1, ME2, and ME3) are involved in cellular energy regulation, redox homeostasis, and biosynthetic processes, through the production of pyruvate and reducing agent NAD(P)H. Recent studies have implicated the third and least well-characterized isoform, mitochondrial NADP+-dependent malic enzyme 3 (ME3), as a therapeutic target for pancreatic cancers. Here, we utilized an integrated structure approach to determine the structures of ME3 in various ligand-binding states at near-atomic resolutions. ME3 is captured in the open form existing as a stable tetramer and its dynamic Domain C is critical for activity. Catalytic assay results reveal that ME3 is a non-allosteric enzyme and does not require modulators for activity while structural analysis suggests that the inner stability of ME3 Domain A relative to ME2 disables allostery in ME3. With structural information available for all three malic enzymes, the foundation has been laid to understand the structural and biochemical differences of these enzymes and could aid in the development of specific malic enzyme small molecule drugs.
Collapse
Affiliation(s)
- Tsehai A.J. Grell
- Structural and Protein Sciences, Janssen Research and Development, LLC, Spring House, Pennsylvania 19477, United States
| | - Mark Mason
- Structural and Protein Sciences, Janssen Research and Development, LLC, Spring House, Pennsylvania 19477, United States
| | - Aaron A. Thompson
- Structural and Protein Sciences, Janssen Research and Development, LLC, San Diego, California 92121, United States
| | | | - Daniel Riley
- Lead Discovery and Molecular Pharmacology, Janssen Research and Development, LLC, Spring House, Pennsylvania 19477, United States
| | - Michelle V. Wagner
- Emerging Science Initiative, Janssen Research and Development, LLC, San Diego, California 92121, United States
| | - Ruth Steele
- Structural and Protein Sciences, Janssen Research and Development, LLC, Spring House, Pennsylvania 19477, United States
| | - Rodrigo F. Ortiz-Meoz
- Lead Discovery and Molecular Pharmacology, Janssen Research and Development, LLC, Spring House, Pennsylvania 19477, United States
| | - Jay Wadia
- Emerging Science Initiative, Janssen Research and Development, LLC, San Diego, California 92121, United States
| | - Paul L. Shaffer
- Structural and Protein Sciences, Janssen Research and Development, LLC, Spring House, Pennsylvania 19477, United States
| | - Gary Tresadern
- Computational Chemistry, Janssen Research and Development, LLC, Beerse, B2340, Belgium
| | - Sujata Sharma
- Structural and Protein Sciences, Janssen Research and Development, LLC, Spring House, Pennsylvania 19477, United States,Structural and Protein Sciences, Janssen Research and Development, LLC, San Diego, California 92121, United States,Corresponding author.
| | - Xiaodi Yu
- Structural and Protein Sciences, Janssen Research and Development, LLC, Spring House, Pennsylvania 19477, United States,Corresponding author.
| |
Collapse
|
24
|
Yoshida T, Kawabe T, Cantley LC, Lyssiotis CA. Discovery and Characterization of a Novel Allosteric Small-Molecule Inhibitor of NADP +-Dependent Malic Enzyme 1. Biochemistry 2022; 61:1548-1553. [PMID: 35819845 PMCID: PMC9352307 DOI: 10.1021/acs.biochem.2c00123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
NADP+-dependent malic enzyme 1 (ME1) decarboxylates
malate to form pyruvate and NADPH in the cytoplasm, where it mediates
diverse biological functions related to the generation of lipids and
other cellular building blocks. As such, ME1 has been implicated in
the progression of cancers and has received attention as a promising
drug target. Here we report the identification of a novel small-molecule
inhibitor of ME1, designated AS1134900. AS1134900 is highly selective
for ME1 compared with ME2 and uncompetitively inhibits ME1 activity
in the presence of its substrates NADP+ and malate. In
addition, X-ray crystal structure analysis of the enzyme–inhibitor
complex revealed that AS1134900 binds outside the ME1 active site
in a novel allosteric site. Structural comparison of the ME1 quaternary
complex with AS1134900, NADPH, and Mn2+, alongside known
crystal structures of malic enzymes, indicated the determined crystal
ME1–inhibitor complex is in the open form conformation. These
results provide insights and a starting point for further discovery
of drugs that inhibit ME1 activity in cancer cells.
Collapse
Affiliation(s)
- Tomohiro Yoshida
- Business Development, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Tetsuhiro Kawabe
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Lewis C Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, New York 10065, United States
| | - Costas A Lyssiotis
- Department of Molecular & Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
25
|
Trovão M, Schüler LM, Machado A, Bombo G, Navalho S, Barros A, Pereira H, Silva J, Freitas F, Varela J. Random Mutagenesis as a Promising Tool for Microalgal Strain Improvement towards Industrial Production. Mar Drugs 2022; 20:440. [PMID: 35877733 PMCID: PMC9318807 DOI: 10.3390/md20070440] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
Microalgae have become a promising novel and sustainable feedstock for meeting the rising demand for food and feed. However, microalgae-based products are currently hindered by high production costs. One major reason for this is that commonly cultivated wildtype strains do not possess the robustness and productivity required for successful industrial production. Several strain improvement technologies have been developed towards creating more stress tolerant and productive strains. While classical methods of forward genetics have been extensively used to determine gene function of randomly generated mutants, reverse genetics has been explored to generate specific mutations and target phenotypes. Site-directed mutagenesis can be accomplished by employing different gene editing tools, which enable the generation of tailor-made genotypes. Nevertheless, strategies promoting the selection of randomly generated mutants avoid the introduction of foreign genetic material. In this paper, we review different microalgal strain improvement approaches and their applications, with a primary focus on random mutagenesis. Current challenges hampering strain improvement, selection, and commercialization will be discussed. The combination of these approaches with high-throughput technologies, such as fluorescence-activated cell sorting, as tools to select the most promising mutants, will also be discussed.
Collapse
Affiliation(s)
- Mafalda Trovão
- Allmicroalgae Natural Products S.A., R&D Department, Rua 25 de Abril s/n, 2445-413 Pataias, Portugal; (M.T.); (A.M.); (A.B.); (J.S.)
- GreenCoLab—Associação Oceano Verde, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (L.M.S.); (G.B.); (S.N.); (H.P.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal;
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Lisa M. Schüler
- GreenCoLab—Associação Oceano Verde, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (L.M.S.); (G.B.); (S.N.); (H.P.)
| | - Adriana Machado
- Allmicroalgae Natural Products S.A., R&D Department, Rua 25 de Abril s/n, 2445-413 Pataias, Portugal; (M.T.); (A.M.); (A.B.); (J.S.)
| | - Gabriel Bombo
- GreenCoLab—Associação Oceano Verde, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (L.M.S.); (G.B.); (S.N.); (H.P.)
| | - Sofia Navalho
- GreenCoLab—Associação Oceano Verde, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (L.M.S.); (G.B.); (S.N.); (H.P.)
| | - Ana Barros
- Allmicroalgae Natural Products S.A., R&D Department, Rua 25 de Abril s/n, 2445-413 Pataias, Portugal; (M.T.); (A.M.); (A.B.); (J.S.)
| | - Hugo Pereira
- GreenCoLab—Associação Oceano Verde, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (L.M.S.); (G.B.); (S.N.); (H.P.)
| | - Joana Silva
- Allmicroalgae Natural Products S.A., R&D Department, Rua 25 de Abril s/n, 2445-413 Pataias, Portugal; (M.T.); (A.M.); (A.B.); (J.S.)
| | - Filomena Freitas
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal;
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - João Varela
- GreenCoLab—Associação Oceano Verde, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (L.M.S.); (G.B.); (S.N.); (H.P.)
- CCMAR—Centre of Marine Sciences, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
26
|
Fazili ABA, Shah AM, Albeshr MF, Naz T, Dar MA, Yang W, Garre V, Fazili KM, Bhat EA, Song Y. Overexpression of the Mitochondrial Malic Enzyme Genes (malC and malD) Improved the Lipid Accumulation in Mucor circinelloides WJ11. Front Microbiol 2022; 13:919364. [PMID: 35814694 PMCID: PMC9260706 DOI: 10.3389/fmicb.2022.919364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/30/2022] [Indexed: 11/18/2022] Open
Abstract
Mucor circinelloides serves as a model organism to investigate the lipid metabolism in oleaginous microorganisms. It is considered as an important producer of γ-linolenic acid (GLA) that has vital medicinal benefits. In this study, we used WJ11, a high lipid-producing strain of M. circinelloides (36% w/w lipid, cell dry weight, CDW), to examine the role in lipid accumulation of two mitochondrial malic enzyme (ME) genes malC and malD. The homologous overexpression of both malC and malD genes enhanced the total lipid content of WJ11 by 41.16 and 32.34%, respectively. In parallel, the total content of GLA was enhanced by 16.73 and 46.76% in malC and malD overexpressing strains, respectively, because of the elevation of total lipid content. The fact that GLA content was enhanced more in the strain with lower lipid content increase and vice versa, indicated that engineering of mitochondrial MEs altered the fatty acid profile. Our results reveal that mitochondrial ME plays an important role in lipid metabolism and suggest that future approaches may involve simultaneous overexpression of distinct ME genes to boost lipid accumulation even further.
Collapse
Affiliation(s)
- Abu Bakr Ahmad Fazili
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, China
| | - Aabid Manzoor Shah
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, China
| | | | - Tahira Naz
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, China
| | | | - Wu Yang
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, China
| | - Victoriano Garre
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | | | - Eijaz Ahmed Bhat
- Centre de Biologie Structurale (CBS), Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Yuanda Song
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, China
- *Correspondence: Yuanda Song,
| |
Collapse
|
27
|
Sano N, Lounifi I, Cueff G, Collet B, Clément G, Balzergue S, Huguet S, Valot B, Galland M, Rajjou L. Multi-Omics Approaches Unravel Specific Features of Embryo and Endosperm in Rice Seed Germination. FRONTIERS IN PLANT SCIENCE 2022; 13:867263. [PMID: 35755645 PMCID: PMC9225960 DOI: 10.3389/fpls.2022.867263] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
Seed germination and subsequent seedling growth affect the final yield and quality of the crop. Seed germination is defined as a series of processes that begins with water uptake by a quiescent dry seed and ends with the elongation of embryonic axis. Rice is an important cereal crop species, and during seed germination, two tissues function in a different manner; the embryo grows into a seedling as the next generation and the endosperm is responsible for nutritional supply. Toward understanding the integrated roles of each tissue at the transcriptional, translational, and metabolic production levels during germination, an exhaustive "multi-omics" analysis was performed by combining transcriptomics, label-free shotgun proteomics, and metabolomics on rice germinating embryo and endosperm, independently. Time-course analyses of the transcriptome and metabolome in germinating seeds revealed a major turning point in the early phase of germination in both embryo and endosperm, suggesting that dramatic changes begin immediately after water imbibition in the rice germination program at least at the mRNA and metabolite levels. In endosperm, protein profiles mostly showed abundant decreases corresponding to 90% of the differentially accumulated proteins. An ontological classification revealed the shift from the maturation to the germination process where over-represented classes belonged to embryonic development and cellular amino acid biosynthetic processes. In the embryo, 19% of the detected proteins are differentially accumulated during germination. Stress response, carbohydrate, fatty acid metabolism, and transport are the main functional classes representing embryo proteome change. Moreover, proteins specific to the germinated state were detected by both transcriptomic and proteomic approaches and a major change in the network operating during rice germination was uncovered. In particular, concomitant changes of hormonal metabolism-related proteins (GID1L2 and CNX1) implicated in GAs and ABA metabolism, signaling proteins, and protein turnover events emphasized the importance of such biological networks in rice seeds. Using metabolomics, we highlighted the importance of an energetic supply in rice seeds during germination. In both embryo and endosperm, starch degradation, glycolysis, and subsequent pathways related to these cascades, such as the aspartate-family pathway, are activated during germination. A relevant number of accumulated proteins and metabolites, especially in embryos, testifies the pivotal role of energetic supply in the preparation of plant growth. This article summarizes the key genetic pathways in embryo and endosperm during rice seed germination at the transcriptional, translational, and metabolite levels and thereby, emphasizes the value of combined multi-omics approaches to uncover the specific feature of tissues during germination.
Collapse
Affiliation(s)
- Naoto Sano
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Imen Lounifi
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
- MBCC Group, Master Builders Construction Chemical, Singapore, Singapore
| | - Gwendal Cueff
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Boris Collet
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Gilles Clément
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Sandrine Balzergue
- Université Paris-Saclay, CNRS, INRAE, Univ Evry, Institute of Plant Sciences Paris-Saclay (IPS2), Orsay, France
- IRHS-UMR1345, Université d'Angers, INRAE, Institut Agro, SFR 4207 QuaSaV, Beaucouzé, France
| | - Stéphanie Huguet
- Université Paris-Saclay, CNRS, INRAE, Univ Evry, Institute of Plant Sciences Paris-Saclay (IPS2), Orsay, France
| | - Benoît Valot
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, GQE - Le Moulon, PAPPSO, Plateforme d'Analyse de Proteomique Paris-Sud-Ouest, Gif-sur-Yvette, France
- Chrono-Environnement Research Team UMR/CNRS-6249, Bourgogne-Franche-Comté University, Besançon, France
| | - Marc Galland
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Loïc Rajjou
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| |
Collapse
|
28
|
Hanau S, Helliwell JR. 6-Phosphogluconate dehydrogenase and its crystal structures. Acta Crystallogr F Struct Biol Commun 2022; 78:96-112. [PMID: 35234135 PMCID: PMC8900737 DOI: 10.1107/s2053230x22001091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/31/2022] [Indexed: 11/10/2022] Open
Abstract
6-Phosphogluconate dehydrogenase (6PGDH; EC 1.1.1.44) catalyses the oxidative decarboxylation of 6-phosphogluconate to ribulose 5-phosphate in the context of the oxidative part of the pentose phosphate pathway. Depending on the species, it can be a homodimer or a homotetramer. Oligomerization plays a functional role not only because the active site is at the interface between subunits but also due to the interlocking tail-modulating activity, similar to that of isocitrate dehydrogenase and malic enzyme, which catalyse a similar type of reaction. Since the pioneering crystal structure of sheep liver 6PGDH, which allowed motifs common to the β-hydroxyacid dehydrogenase superfamily to be recognized, several other 6PGDH crystal structures have been solved, including those of ternary complexes. These showed that more than one conformation exists, as had been suggested for many years from enzyme studies in solution. It is inferred that an asymmetrical conformation with a rearrangement of one of the two subunits underlies the homotropic cooperativity. There has been particular interest in the presence or absence of sulfate during crystallization. This might be related to the fact that this ion, which is a competitive inhibitor that binds in the active site, can induce the same 6PGDH configuration as in the complexes with physiological ligands. Mutagenesis, inhibitors, kinetic and binding studies, post-translational modifications and research on the enzyme in cancer cells have been complementary to the crystallographic studies. Computational modelling and new structural studies will probably help to refine the understanding of the functioning of this enzyme, which represents a promising therapeutic target in immunity, cancer and infective diseases. 6PGDH also has applied-science potential as a biosensor or a biobattery. To this end, the enzyme has been efficiently immobilized on specific polymers and nanoparticles. This review spans the 6PGDH literature and all of the 6PGDH crystal structure data files held by the Protein Data Bank.
Collapse
Affiliation(s)
- Stefania Hanau
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Borsari 46, Ferrara, Italy
| | - John R. Helliwell
- Department of Chemistry, University of Manchester, Manchester M13 9PL, United Kingdom
| |
Collapse
|
29
|
Hüdig M, Tronconi MA, Zubimendi JP, Sage TL, Poschmann G, Bickel D, Gohlke H, Maurino VG. Respiratory and C4-photosynthetic NAD-malic enzyme coexist in bundle sheath cell mitochondria and evolved via association of differentially adapted subunits. THE PLANT CELL 2022; 34:597-615. [PMID: 34734993 PMCID: PMC8773993 DOI: 10.1093/plcell/koab265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/26/2021] [Indexed: 05/29/2023]
Abstract
In plant mitochondria, nicotinamide adenine dinucleotide-malic enzyme (NAD-ME) has a housekeeping function in malate respiration. In different plant lineages, NAD-ME was independently co-opted in C4 photosynthesis. In the C4 Cleome species, Gynandropsis gynandra and Cleome angustifolia, all NAD-ME genes (NAD-MEα, NAD-MEβ1, and NAD-MEβ2) were affected by C4 evolution and are expressed at higher levels than their orthologs in the C3 species Tarenaya hassleriana. In T. hassleriana, the NAD-ME housekeeping function is performed by two heteromers, NAD-MEα/β1 and NAD-MEα/β2, with similar biochemical properties. In both C4 species, this role is restricted to NAD-MEα/β2. In the C4 species, NAD-MEα/β1 is exclusively present in the leaves, where it accounts for most of the enzymatic activity. Gynandropsis gynandra NAD-MEα/β1 (GgNAD-MEα/β1) exhibits high catalytic efficiency and is differentially activated by the C4 intermediate aspartate, confirming its role as the C4-decarboxylase. During C4 evolution, NAD-MEβ1 lost its catalytic activity; its contribution to the enzymatic activity results from a stabilizing effect on the associated α-subunit and the acquisition of regulatory properties. We conclude that in bundle sheath cell mitochondria of C4 species, the functions of NAD-ME as C4 photosynthetic decarboxylase and as a housekeeping enzyme coexist and are performed by isoforms that combine the same α-subunit with differentially adapted β-subunits.
Collapse
Affiliation(s)
- Meike Hüdig
- Molekulare Pflanzenphysiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Kirschallee, Bonn 53115, Germany
| | - Marcos A Tronconi
- Centro de Estudios Fotosintéticos y Bioquímicos, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina
| | - Juan P Zubimendi
- Centro de Estudios Fotosintéticos y Bioquímicos, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina
| | - Tammy L Sage
- Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | - Gereon Poschmann
- Molecular Proteomics Laboratory, Biomedical Research Centre (BMFZ) & Institute of Molecular Medicine, Proteome Research, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - David Bickel
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - Holger Gohlke
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
- John von Neumann Institute for Computing (NIC), Jülich Supercomputing Centre (JSC), Institute of Biological Information Processing (IBI-7: Structural Biochemistry) & Institute of Bio- and Geosciences (IBG-4: Bioinformatics), Forschungszentrum Jülich GmbH, Jülich 52425, Germany
| | - Veronica G Maurino
- Molekulare Pflanzenphysiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Kirschallee, Bonn 53115, Germany
| |
Collapse
|
30
|
Chen LL, Wang WJ. p53 regulates lipid metabolism in cancer. Int J Biol Macromol 2021. [DOI: https://doi.org/10.1016/j.ijbiomac.2021.09.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
31
|
Chen LL, Wang WJ. p53 regulates lipid metabolism in cancer. Int J Biol Macromol 2021; 192:45-54. [PMID: 34619274 DOI: 10.1016/j.ijbiomac.2021.09.188] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/22/2021] [Accepted: 09/28/2021] [Indexed: 02/07/2023]
Abstract
Reprogrammed cell metabolism is a well-accepted hallmark of cancer. Metabolism changes provide energy and precursors for macromolecule biosynthesis to satisfy the survival needs of cancer cells. The specific changes in different aspects of lipid metabolism in cancer cells have been focused in recent years. These changes can affect cell growth, proliferation, differentiation and motility through affecting membranes synthesis, energy homeostasis and cell signaling. The tumor suppressor p53 plays vital roles in the control of cell proliferation, senescence, DNA repair, and cell death in cancer through various transcriptional and non-transcriptional activities. Accumulating evidences indicate that p53 also regulates cellular metabolism, which appears to contribute to its tumor suppressive functions. Particularly the role of p53 in regulating lipid metabolism has gained more and more attention in recent decades. In this review, we summarize recent advances in the function of p53 on lipid metabolism in cancer. Further understanding and research on the role of p53 in lipid metabolism regulation will provide a potential therapeutic window for cancer treatment.
Collapse
Affiliation(s)
- Ling-Li Chen
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Wen-Jun Wang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China.
| |
Collapse
|
32
|
Gelsinger DR, Reddy R, Whittington K, Debic S, DiRuggiero J. Post-transcriptional regulation of redox homeostasis by the small RNA SHOxi in haloarchaea. RNA Biol 2021; 18:1867-1881. [PMID: 33522404 PMCID: PMC8583180 DOI: 10.1080/15476286.2021.1874717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 11/13/2022] Open
Abstract
While haloarchaea are highly resistant to oxidative stress, a comprehensive understanding of the processes regulating this remarkable response is lacking. Oxidative stress-responsive small non-coding RNAs (sRNAs) have been reported in the model archaeon, Haloferax volc anii, but targets and mechanisms have not been elucidated. Using a combination of high throughput and reverse molecular genetic approaches, we elucidated the functional role of the most up-regulated intergenic sRNA during oxidative stress in H. volcanii, named Small RNA in Haloferax Oxidative Stress (SHOxi). SHOxi was predicted to form a stable secondary structure with a conserved stem-loop region as the potential binding site for trans-targets. NAD-dependent malic enzyme mRNA, identified as a putative target of SHOxi, interacted directly with a putative 'seed' region within the predicted stem loop of SHOxi. Malic enzyme catalyzes the oxidative decarboxylation of malate into pyruvate using NAD+ as a cofactor. The destabilization of malic enzyme mRNA, and the decrease in the NAD+/NADH ratio, resulting from the direct RNA-RNA interaction between SHOxi and its trans-target was essential for the survival of H. volcanii to oxidative stress. These findings indicate that SHOxi likely regulates redox homoeostasis during oxidative stress by the post-transcriptional destabilization of malic enzyme mRNA. SHOxi-mediated regulation provides evidence that the fine-tuning of metabolic cofactors could be a core strategy to mitigate damage from oxidative stress and confer resistance. This study is the first to establish the regulatory effects of sRNAs on mRNAs during the oxidative stress response in Archaea.
Collapse
Affiliation(s)
| | - Rahul Reddy
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Sara Debic
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Jocelyne DiRuggiero
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Zhang S, Cheng ZM, Yu JL, Lu K, Xu SJ, Lu Y, Liu T, Xia BJ, Huang Z, Zhao XY, He W, Li JX, Cao W, Huang Y, Wang L, Zeng Z, Zou X, Liu R, Zhang YS, Wu XP, Jiang TP, Zhou S. Malic enzyme 2 promotes the progression of hepatocellular carcinoma via increasing triglyceride production. Cancer Med 2021; 10:6795-6806. [PMID: 34427987 PMCID: PMC8495273 DOI: 10.1002/cam4.4209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/13/2021] [Accepted: 07/02/2021] [Indexed: 12/28/2022] Open
Abstract
The incidence and mortality of hepatocellular carcinoma (HCC) are gradually increasing during the past years. Recently, some studies have reported that malic enzyme (ME) plays an important role in cancer development, while the involvement of ME2 in HCC remains still undetermined. Here, we demonstrated that ME2 played an oncogenic role in HCC. ME2 was overexpressed in HCC tissues. TCGA database showed that the ME2 transcript level was inversely associated with the survival of HCC patients. Loss‐of‐function and gain‐of‐function assays showed that ME2 promoted HCC cell growth and migration. Furthermore, the xenografted tumorigenesis of MHCC97H cells was retarded by ME2 knockdown. ME2 silencing also suppressed the cell cycle process and induced apoptosis. Mechanistically, ME2 potentiated triglyceride synthesis, inhibition of which suppressed the proliferation and migration. We propose that ME2 promotes HCC progression by increasing triglyceride production.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China.,Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhi-Mei Cheng
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jia-Li Yu
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Kai Lu
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Sheng-Jie Xu
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Yuan Lu
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Ting Liu
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Bai-Juan Xia
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Zhi Huang
- Department of Radiology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xu-Ya Zhao
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Wei He
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Jun-Xiang Li
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Wei Cao
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Yu Huang
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Ling Wang
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Zhu Zeng
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Xun Zou
- Department of Radiology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Rong Liu
- Department of Interventional Radiology, First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, China
| | - Yu-Sui Zhang
- Department of Interventional Radiology, First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, China
| | - Xiao-Ping Wu
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tian-Peng Jiang
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shi Zhou
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China.,Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
34
|
Hofmann J, Bitew MA, Kuba M, De Souza DP, Newton HJ, Sansom FM. Characterisation of putative lactate synthetic pathways of Coxiella burnetii. PLoS One 2021; 16:e0255925. [PMID: 34388185 PMCID: PMC8362950 DOI: 10.1371/journal.pone.0255925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/26/2021] [Indexed: 11/19/2022] Open
Abstract
The zoonotic pathogen Coxiella burnetii, the causative agent of the human disease Q fever, is an ever-present danger to global public health. Investigating novel metabolic pathways necessary for C. burnetii to replicate within its unusual intracellular niche may identify new therapeutic targets. Recent studies employing stable isotope labelling established the ability of C. burnetii to synthesize lactate, despite the absence of an annotated synthetic pathway on its genome. A noncanonical lactate synthesis pathway could provide a novel anti-Coxiella target if it is essential for C. burnetii pathogenesis. In this study, two C. burnetii proteins, CBU1241 and CBU0823, were chosen for analysis based on their similarities to known lactate synthesizing enzymes. Recombinant GST-CBU1241, a putative malate dehydrogenase (MDH), did not produce measurable lactate in in vitro lactate dehydrogenase (LDH) activity assays and was confirmed to function as an MDH. Recombinant 6xHis-CBU0823, a putative NAD+-dependent malic enzyme, was shown to have both malic enzyme activity and MDH activity, however, did not produce measurable lactate in either LDH or malolactic enzyme activity assays in vitro. To examine potential lactate production by CBU0823 more directly, [13C]glucose labelling experiments compared label enrichment within metabolic pathways of a cbu0823 transposon mutant and the parent strain. No difference in lactate production was observed, but the loss of CBU0823 significantly reduced 13C-incorporation into glycolytic and TCA cycle intermediates. This disruption to central carbon metabolism did not have any apparent impact on intracellular replication within THP-1 cells. This research provides new information about the mechanism of lactate biosynthesis within C. burnetii, demonstrating that CBU1241 is not multifunctional, at least in vitro, and that CBU0823 also does not synthesize lactate. Although critical for normal central carbon metabolism of C. burnetii, loss of CBU0823 did not significantly impair replication of the bacterium inside cells.
Collapse
Affiliation(s)
- Janine Hofmann
- Faculty of Veterinary and Agricultural Sciences, Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, University of Melbourne, Parkville, Victoria, Australia
| | - Mebratu A. Bitew
- Department of Pathology, Microbiology and Immunology, University of California, Davis, California, United States of America
| | - Miku Kuba
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - David P. De Souza
- Metabolomics Australia, The Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Hayley J. Newton
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Fiona M. Sansom
- Faculty of Veterinary and Agricultural Sciences, Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
35
|
Mercaldi GF, Eufrásio AG, Ranzani AT, do Nascimento Faria J, Mota SGR, Fagundes M, Bruder M, Cordeiro AT. Trypanosoma cruzi Malic Enzyme Is the Target for Sulfonamide Hits from the GSK Chagas Box. ACS Infect Dis 2021; 7:2455-2471. [PMID: 34279922 DOI: 10.1021/acsinfecdis.1c00231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chagas disease, an infectious condition caused by Trypanosoma cruzi, lacks treatment with drugs with desired efficacy and safety profiles. To address this unmet medical need, a set of trypanocidal compounds were identified through a large multicenter phenotypic-screening initiative and assembled in the GSK Chagas Box. In the present work, we report the screening of the Chagas Box against T. cruzi malic enzymes (MEs) and the identification of three potent inhibitors of its cytosolic isoform (TcMEc). One of these compounds, TCMDC-143108 (1), came out as a nanomolar inhibitor of TcMEc, and 14 new derivatives were synthesized and tested for target inhibition and efficacy against the parasite. Moreover, we determined the crystallographic structures of TcMEc in complex with TCMDC-143108 (1) and six derivatives, revealing the allosteric inhibition site and the determinants of specificity. Our findings connect phenotypic hits from the Chagas Box to a relevant metabolic target in the parasite, providing data to foster new structure-activity guided hit optimization initiatives.
Collapse
Affiliation(s)
- Gustavo F. Mercaldi
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
| | - Amanda G. Eufrásio
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
- Faculty of Pharmaceutic Sciences, University of Campinas, Campinas, SP 13083-871, Brazil
| | - Americo T. Ranzani
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
- Institute of Biology, University of Campinas, Campinas, SP 13083-970, Brazil
| | - Jessica do Nascimento Faria
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
| | - Sabrina G. R. Mota
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
- Institute of Biology, University of Campinas, Campinas, SP 13083-970, Brazil
| | - Michelle Fagundes
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
| | - Marjorie Bruder
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
| | - Artur T. Cordeiro
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
- Faculty of Pharmaceutic Sciences, University of Campinas, Campinas, SP 13083-871, Brazil
| |
Collapse
|
36
|
Prochownik EV, Wang H. The Metabolic Fates of Pyruvate in Normal and Neoplastic Cells. Cells 2021; 10:cells10040762. [PMID: 33808495 PMCID: PMC8066905 DOI: 10.3390/cells10040762] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 02/06/2023] Open
Abstract
Pyruvate occupies a central metabolic node by virtue of its position at the crossroads of glycolysis and the tricarboxylic acid (TCA) cycle and its production and fate being governed by numerous cell-intrinsic and extrinsic factors. The former includes the cell’s type, redox state, ATP content, metabolic requirements and the activities of other metabolic pathways. The latter include the extracellular oxygen concentration, pH and nutrient levels, which are in turn governed by the vascular supply. Within this context, we discuss the six pathways that influence pyruvate content and utilization: 1. The lactate dehydrogenase pathway that either converts excess pyruvate to lactate or that regenerates pyruvate from lactate for use as a fuel or biosynthetic substrate; 2. The alanine pathway that generates alanine and other amino acids; 3. The pyruvate dehydrogenase complex pathway that provides acetyl-CoA, the TCA cycle’s initial substrate; 4. The pyruvate carboxylase reaction that anaplerotically supplies oxaloacetate; 5. The malic enzyme pathway that also links glycolysis and the TCA cycle and generates NADPH to support lipid bio-synthesis; and 6. The acetate bio-synthetic pathway that converts pyruvate directly to acetate. The review discusses the mechanisms controlling these pathways, how they cross-talk and how they cooperate and are regulated to maximize growth and achieve metabolic and energetic harmony.
Collapse
Affiliation(s)
- Edward V. Prochownik
- Division of Hematology/Oncology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA;
- The Department of Microbiology and Molecular Genetics, UPMC, Pittsburgh, PA 15213, USA
- The Hillman Cancer Center, UPMC, Pittsburgh, PA 15213, USA
- The Pittsburgh Liver Research Center, Pittsburgh, PA 15260, USA
- Correspondence: ; Tel.: +1-(412)-692-6795
| | - Huabo Wang
- Division of Hematology/Oncology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA;
| |
Collapse
|
37
|
Emami NK, Cauble RN, Dhamad AE, Greene ES, Coy CS, Velleman SG, Orlowski S, Anthony N, Bedford M, Dridi S. Hypoxia further exacerbates woody breast myopathy in broilers via alteration of satellite cell fate. Poult Sci 2021; 100:101167. [PMID: 34091348 PMCID: PMC8182261 DOI: 10.1016/j.psj.2021.101167] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/09/2021] [Accepted: 03/19/2021] [Indexed: 11/30/2022] Open
Abstract
Woody breast (WB) condition has created a variety of challenges for the global poultry industry. To date, there are no effective treatments or preventative measures due to its unknown (undefined) etiology. Several potential mechanisms including oxidative stress, fiber-type switching, cellular damage, and altered intracellular calcium levels have been proposed to play a key role in the progression of the WB myopathy. In a previous study, we have shown that WB is associated with hypoxia-like status and dysregulated oxygen homeostasis. As satellite cells (SC) play a pivotal role in muscle fiber repair and remodeling under stress conditions, we undertook the present study to determine satellite cell fate in WB-affected birds when reared in either normoxic or hypoxic conditions. Modern random bred broilers from 2015 (n = 200) were wing banded and reared under standard brooding practices for the first 2 wk post-hatch. At 15 d, chicks were divided in 2 body weight-matched groups and reared to 6 wk in either control local altitude or hypobaric chambers with simulated altitude of 6,000 ft. Birds were provided ad libitum access to water and feed, according to the Cobb recommendations. At 6 wk of age, birds were processed and scored for WB, and breast samples were collected from WB-affected and unaffected birds for molecular analyses (n = 10/group). SCs were isolated from normal breast muscle, cultured in vitro, and exposed to normoxia or hypoxia for 2 h. The expression of target genes was determined by qPCR using 2−∆∆Ct method. Protein distribution and expression were determined by immunofluorescence staining and immunoblot, respectively. Data were analyzed by the Student's t test with significance set at P < 0.05. Multiple satellite cell markers, myogenic factor (Myf)-5 and paired box (PAX)-7 were significantly decreased at the mRNA and protein levels in the breast muscle from WB-affected birds compared to their unaffected counterparts. Lipogenic-and adipogenic-associated factors (acetyl-CoA carboxylase, ACCα; fatty acid synthase, FASN, malic enzyme, ME; and ATP citrate lyase, ACLY) were activated in WB-affected birds. These data were supported by an in vitro study where hypoxia decreased the expression of Myf5 and Pax7, and increased that of ACCα, FASN, ME, and ACLY. Together, these data indicate that under hypoxic condition, SC change fate by switching from a myogenic to an adipogenic program, which explains at least partly, the etiology of the WB myopathy.
Collapse
Affiliation(s)
- Nima K Emami
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, 1260 W. Maple Street, Fayetteville 72701, USA
| | - Reagan N Cauble
- Department of Animal Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Ahmed E Dhamad
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Elizabeth S Greene
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, 1260 W. Maple Street, Fayetteville 72701, USA
| | - Cynthia S Coy
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Sandra G Velleman
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Sara Orlowski
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, 1260 W. Maple Street, Fayetteville 72701, USA
| | - Nicholas Anthony
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, 1260 W. Maple Street, Fayetteville 72701, USA
| | | | - Sami Dridi
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, 1260 W. Maple Street, Fayetteville 72701, USA.
| |
Collapse
|
38
|
Single nucleotide variants lead to dysregulation of the human mitochondrial NAD(P) +-dependent malic enzyme. iScience 2021; 24:102034. [PMID: 33554057 PMCID: PMC7847962 DOI: 10.1016/j.isci.2021.102034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 11/16/2020] [Accepted: 12/31/2020] [Indexed: 11/23/2022] Open
Abstract
Human mitochondrial NAD(P)+-dependent malic enzyme (ME2) is well recognized to associate with cancer cell metabolism, and the single nucleotide variants (SNVs) of ME2 may play a role in enzyme regulation. Here we reported that the SNVs of ME2 occurring in the allosteric sites lead to inactivation or overactivation of ME2. Two ME2-SNVs, ME2_R67Q and ME2-R484W, that demonstrated inactivating or overactivating enzyme activities of ME2, respectively, have different impact toward the cells. The cells with overactivating SNV enzyme, ME2_R484W, grow more rapidly and are more resistant to cellular senescence than the cells with wild-type or inactivating SNV enzyme, ME2_R67Q. Crystal structures of these two ME2-SNVs reveal that ME2_R67Q was an inactivating "dead form," and ME2_R484W was an overactivating "closed form" of the enzyme. The resolved ME2-SNV structures provide a molecular basis to explain the abnormal kinetic properties of these SNV enzymes.
Collapse
|
39
|
Enzyme-based amperometric biosensors for malic acid - A review. Anal Chim Acta 2021; 1156:338218. [PMID: 33781460 DOI: 10.1016/j.aca.2021.338218] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/18/2022]
Abstract
Malic acid is a key flavour component of many fruits and vegetables. There is significant interest in technologies for monitoring its concentration, particularly in winemaking. In this review we systematically and comprehensively chart progress in the development of enzyme-based amperometric biosensors for malic acid. We summarise the components and analytical parameters of malic acid sensors that have been reported over the past four decades, discussing their merits and pitfalls in terms of accuracy, sensitivity, linear range, response time and stability. We discuss how advances in electrode materials, electron mediators and the use of coupled enzymes have improved sensitivity and minimised interference, but also uncover a trade-off between sensitivity and linear range. A particular focus of our review is the three types of malate oxidoreductase enzyme that have been used in malic acid biosensors. We describe their different properties and conclude that identifying and/or engineering superior alternatives will be a key future direction for improving the commercial utility of malic acid biosensors.
Collapse
|
40
|
Burley KH, Cuthbert BJ, Basu P, Newcombe J, Irimpan EM, Quechol R, Foik IP, Mobley DL, Beste DJV, Goulding CW. Structural and Molecular Dynamics of Mycobacterium tuberculosis Malic Enzyme, a Potential Anti-TB Drug Target. ACS Infect Dis 2021; 7:174-188. [PMID: 33356117 DOI: 10.1021/acsinfecdis.0c00735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tuberculosis (TB) is the most lethal bacterial infectious disease worldwide. It is notoriously difficult to treat, requiring a cocktail of antibiotics administered over many months. The dense, waxy outer membrane of the TB-causing agent, Mycobacterium tuberculosis (Mtb), acts as a formidable barrier against uptake of antibiotics. Subsequently, enzymes involved in maintaining the integrity of the Mtb cell wall are promising drug targets. Recently, we demonstrated that Mtb lacking malic enzyme (MEZ) has altered cell wall lipid composition and attenuated uptake by macrophages. These results suggest that MEZ contributes to lipid biosynthesis by providing reductants in the form of NAD(P)H. Here, we present the X-ray crystal structure of MEZ to 3.6 Å. We use biochemical assays to demonstrate MEZ is dimeric in solution and to evaluate the effects of pH and allosteric regulators on its kinetics and thermal stability. To assess the interactions between MEZ and its substrate malate and cofactors, Mn2+ and NAD(P)+, we ran a series of molecular dynamics (MD) simulations. First, the MD analysis corroborates our empirical observations that MEZ is unusually flexible, which persists even with the addition of substrate and cofactors. Second, the MD simulations reveal that dimeric MEZ subunits alternate between open and closed states, and that MEZ can stably bind its NAD(P)+ cofactor in multiple conformations, including an inactive, compact NAD+ form. Together the structure of MEZ and insights from its dynamics can be harnessed to inform the design of MEZ inhibitors that target Mtb and not human malic enzyme homologues.
Collapse
Affiliation(s)
| | | | - Piyali Basu
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Jane Newcombe
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | | | | | | | | | - Dany J. V. Beste
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | | |
Collapse
|
41
|
Eprintsev AT, Gataullina MO. Kinetic Properties of NADP+-Dependent Decarboxylating Malate Dehydrogenase from Corn Leaves. APPL BIOCHEM MICRO+ 2021. [DOI: 10.1134/s0003683821010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
You D, Du D, Zhao X, Li X, Ying M, Hu X. Mitochondrial malic enzyme 2 promotes breast cancer metastasis via stabilizing HIF-1α under hypoxia. Chin J Cancer Res 2021; 33:308-322. [PMID: 34321828 PMCID: PMC8286887 DOI: 10.21147/j.issn.1000-9604.2021.03.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/22/2021] [Indexed: 12/29/2022] Open
Abstract
Objective α-ketoglutarate (α-KG) is the substrate to hydroxylate collagen and hypoxia-inducible factor-1α (HIF-1α), which are important for cancer metastasis. Previous studies have shown that the upregulation of collagen prolyl 4-hydroxylase in breast cancer cells stabilizes the expression of HIF-1α by depleting α-KG levels. We hypothesized that mitochondrial malic enzyme 2 (ME2) might also affect HIF-1α expression via modulating α-KG levels in breast cancer cells. Methods We evaluated ME2 protein expression in 100 breast cancer patients using immunohistochemistry and correlated with clinicopathological indicators. The effect of ME2 knockout on cancer metastasis was evaluated using an orthotopic breast cancer model. The effect of ME2 knockout or knockdown on the levels of α-KG and HIF-1α proteins in breast cancer cell lines was determined both in vitro and in vivo. Results ME2 was found to be upregulated in the human breast cancerous tissues compared with the matched precancerous tissues (P<0.001). The elevated expression of ME2 was associated with a poor prognosis (P=0.019). ME2 upregulation was also related to lymph node metastasis (P=0.016), pathological staging (P=0.033), and vascular cancer embolus (P=0.014). Also, ME2 knockout significantly inhibited lung metastasisin vivo. In the tumors formed by ME2 knockout cells, the levels of α-KG were significantly increased and collagen hydroxylation level did not change significantly but HIF-1α protein expression was significantly decreased, compared to the control samples. In cell culture, cells with ME2 knockout or knockdown demonstrated significantly higher α-KG levels but significantly lower HIF-1α protein expression than control cells under hypoxia. Exogenous malate and α-KG exerted similar effect on HIF-1α in breast cancer cells to ME2 knockout or knockdown. Additionally, treatment with malate significantly decreased 4T1 breast cancer lung metastasis. ME2 expression was associated with HIF-1α levels in human breast cancer samples (P=0.008). Conclusions Our results provide evidence that upregulation of ME2 is associated with a poor prognosis of breast cancer patients and propose a mechanistic understanding of a link between ME2 and breast cancer metastasis.
Collapse
Affiliation(s)
- Duo You
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Danfeng Du
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory of Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou 450000, China
| | - Xueke Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory of Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou 450000, China
| | - Xinmin Li
- Department of Pathology, Women and Infants Hospital of Zhengzhou, Zhengzhou 450000, China
| | - Minfeng Ying
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xun Hu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| |
Collapse
|
43
|
Tronconi MA, Hüdig M, Schranz ME, Maurino VG. Independent Recruitment of Duplicated β-Subunit-Coding NAD-ME Genes Aided the Evolution of C4 Photosynthesis in Cleomaceae. FRONTIERS IN PLANT SCIENCE 2020; 11:572080. [PMID: 33123181 PMCID: PMC7573226 DOI: 10.3389/fpls.2020.572080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/14/2020] [Indexed: 05/21/2023]
Abstract
In different lineages of C4 plants, the release of CO2 by decarboxylation of a C4 acid near rubisco is catalyzed by NADP-malic enzyme (ME) or NAD-ME, and the facultative use of phosphoenolpyruvate carboxykinase. The co-option of gene lineages during the evolution of C4-NADP-ME has been thoroughly investigated, whereas that of C4-NAD-ME has received less attention. In this work, we aimed at elucidating the mechanism of recruitment of NAD-ME for its function in the C4 pathway by focusing on the eudicot family Cleomaceae. We identified a duplication of NAD-ME in vascular plants that generated the two paralogs lineages: α- and β-NAD-ME. Both gene lineages were retained across seed plants, and their fixation was likely driven by a degenerative process of sub-functionalization, which resulted in a NAD-ME operating primarily as a heteromer of α- and β-subunits. We found most angiosperm genomes maintain a 1:1 β-NAD-ME/α-NAD-ME (β/α) relative gene dosage, but with some notable exceptions mainly due to additional duplications of β-NAD-ME subunits. For example, a significantly high proportion of species with C4-NAD-ME-type photosynthesis have a non-1:1 ratio of β/α. In the Brassicales, we found C4 species with a 2:1 ratio due to a β-NAD-ME duplication (β1 and β2); this was also observed in the C3 Tarenaya hassleriana and Brassica crops. In the independently evolved C4 species, Gynandropsis gynandra and Cleome angustifolia, all three genes were affected by C4 evolution with α- and β1-NAD-ME driven by adaptive selection. In particular, the β1-NAD-MEs possess many differentially substituted amino acids compared with other species and the β2-NAD-MEs of the same species. Five of these amino acids are identically substituted in β1-NAD-ME of G. gynandra and C. angustifolia, two of them were identified as positively selected. Using synteny analysis, we established that β-NAD-ME duplications were derived from ancient polyploidy events and that α-NAD-ME is in a unique syntenic context in both Cleomaceae and Brassicaceae. We discuss our hypotheses for the evolution of NAD-ME and its recruitment for C4 photosynthesis. We propose that gene duplications provided the basis for the recruitment of NAD-ME in C4 Cleomaceae and that all members of the NAD-ME gene family have been adapted to fit the C4-biochemistry. Also, one of the β-NAD-ME gene copies was independently co-opted for its function in the C4 pathway.
Collapse
Affiliation(s)
- Marcos A. Tronconi
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Meike Hüdig
- Abteilung Molekulare Pflanzenphysiologie, Institut für Molekulare Physiologie und Biotechnologie der Pflanzen, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - M. Eric Schranz
- Biosystematics Group, Wageningen University, Wageningen, Netherlands
| | - Veronica G. Maurino
- Abteilung Molekulare Pflanzenphysiologie, Institut für Molekulare Physiologie und Biotechnologie der Pflanzen, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
44
|
Mendes Ferreira A, Mendes-Faia A. The Role of Yeasts and Lactic Acid Bacteria on the Metabolism of Organic Acids during Winemaking. Foods 2020; 9:E1231. [PMID: 32899297 PMCID: PMC7555314 DOI: 10.3390/foods9091231] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 12/31/2022] Open
Abstract
The main role of acidity and pH is to confer microbial stability to wines. No less relevant, they also preserve the color and sensory properties of wines. Tartaric and malic acids are generally the most prominent acids in wines, while others such as succinic, citric, lactic, and pyruvic can exist in minor concentrations. Multiple reactions occur during winemaking and processing, resulting in changes in the concentration of these acids in wines. Two major groups of microorganisms are involved in such modifications: the wine yeasts, particularly strains of Saccharomyces cerevisiae, which carry out alcoholic fermentation; and lactic acid bacteria, which commonly conduct malolactic fermentation. This review examines various such modifications that occur in the pre-existing acids of grape berries and in others that result from this microbial activity as a means to elucidate the link between microbial diversity and wine composition.
Collapse
Affiliation(s)
- Ana Mendes Ferreira
- University of Trás-os-Montes and Alto Douro, 5001-801 Vila Real, Portugal;
- WM&B—Wine Microbiology & Biotechnology Laboratory, Department of Biology and Environment, UTAD, 5001-801 Vila Real, Portugal
- BioISI—Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, 1749-016 Lisboa, Portugal
| | - Arlete Mendes-Faia
- University of Trás-os-Montes and Alto Douro, 5001-801 Vila Real, Portugal;
- WM&B—Wine Microbiology & Biotechnology Laboratory, Department of Biology and Environment, UTAD, 5001-801 Vila Real, Portugal
- BioISI—Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, 1749-016 Lisboa, Portugal
| |
Collapse
|
45
|
Genomic dissection and expression analysis of stress-responsive genes in C4 panicoid models, Setaria italica and Setaria viridis. J Biotechnol 2020; 318:57-67. [DOI: 10.1016/j.jbiotec.2020.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/23/2020] [Accepted: 05/11/2020] [Indexed: 02/02/2023]
|
46
|
Pan X, Fan Z, Chen L, Liu C, Bai F, Wei Y, Tian Z, Dong Y, Shi J, Chen H, Jin Y, Cheng Z, Jin S, Lin J, Wu W. PvrA is a novel regulator that contributes to Pseudomonas aeruginosa pathogenesis by controlling bacterial utilization of long chain fatty acids. Nucleic Acids Res 2020; 48:5967-5985. [PMID: 32406921 PMCID: PMC7293031 DOI: 10.1093/nar/gkaa377] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/19/2022] Open
Abstract
During infection of a host, Pseudomonas aeruginosa orchestrates global gene expression to adapt to the host environment and counter the immune attacks. P. aeruginosa harbours hundreds of regulatory genes that play essential roles in controlling gene expression. However, their contributions to the bacterial pathogenesis remain largely unknown. In this study, we analysed the transcriptomic profile of P. aeruginosa cells isolated from lungs of infected mice and examined the roles of upregulated regulatory genes in bacterial virulence. Mutation of a novel regulatory gene pvrA (PA2957) attenuated the bacterial virulence in an acute pneumonia model. Chromatin immunoprecipitation (ChIP)-Seq and genetic analyses revealed that PvrA directly regulates genes involved in phosphatidylcholine utilization and fatty acid catabolism. Mutation of the pvrA resulted in defective bacterial growth when phosphatidylcholine or palmitic acid was used as the sole carbon source. We further demonstrated that palmitoyl coenzyme A is a ligand for the PvrA, enhancing the binding affinity of PvrA to its target promoters. An arginine residue at position 136 was found to be essential for PvrA to bind palmitoyl coenzyme A. Overall, our results revealed a novel regulatory pathway that controls genes involved in phosphatidylcholine and fatty acid utilization and contributes to the bacterial virulence.
Collapse
Affiliation(s)
- Xiaolei Pan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zheng Fan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lei Chen
- Department of Plant Biology and Ecology, College of Life Science Nankai University, Tianjin 300071 China
| | - Chang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fang Bai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yu Wei
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Zhenyang Tian
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuanyuan Dong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jing Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hao Chen
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhihui Cheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shouguang Jin
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jianping Lin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
47
|
Nichols K, Bannink A, van Baal J, Dijkstra J. Impact of post-ruminally infused macronutrients on bovine mammary gland expression of genes involved in fatty acid synthesis, energy metabolism, and protein synthesis measured in RNA isolated from milk fat. J Anim Sci Biotechnol 2020; 11:53. [PMID: 32477515 PMCID: PMC7238732 DOI: 10.1186/s40104-020-00456-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/01/2020] [Indexed: 11/10/2022] Open
Abstract
Background Characterising the regulation of milk component synthesis in response to macronutrient supply is critical for understanding the implications of nutritional interventions on milk production. Gene expression in mammary gland secretory cells was measured using RNA isolated from milk fat globules from 6 Holstein-Friesian cows receiving 5-d abomasal infusions of saline, essential amino acids (AA), or glucose (GG) or palm olein (LG) without (LAA) or with (HAA) essential AA, according to a 6 × 6 Latin square design. RNA was isolated from milk fat samples collected on d 5 of infusion and subjected to real-time quantitative PCR. We hypothesised that mRNA expression of genes involved in de novo milk fatty acid (FA) synthesis would be differently affected by GG and LG, and that expression of genes regulating transfer of tricarboxylic acid cycle intermediates would increase at the HAA level. We also hypothesised that the HAA level would affect genes regulating endoplasmic reticulum (ER) homeostasis but would not affect genes related to the mechanistic target of rapamycin complex 1 (mTORC1) or the integrated stress response (ISR) network. Results Infusion of GG did not affect de novo milk FA yield but decreased expression of FA synthase (FASN). Infusion of LG decreased de novo FA yield and tended to decrease expression of acetyl-CoA carboxylase 1 (ACC1). The HAA level increased both de novo FA yield and expression of ACC1, and tended to decrease expression of mitochondrial phosphoenolpyruvate carboxykinase (PCK2). mRNA expression of mTORC1 signaling participants was not affected by GG, LG, or AA level. Expression of the ε subunit of the ISR constituent eukaryotic translation initiation factor 2B (EIF2B5) tended to increase at the HAA level, but only in the presence of LG. X-box binding protein 1 (XBP1) mRNA was activated in response to LG and the HAA level. Conclusions Results show that expression of genes involved in de novo FA synthesis responded to glucogenic, lipogenic, and aminogenic substrates, whereas genes regulating intermediate flux through the tricarboxylic acid cycle were not majorly affected. Results also suggest that after 5 d of AA supplementation, milk protein synthesis is supported by enhanced ER biogenesis instead of signaling through the mTORC1 or ISR networks.
Collapse
Affiliation(s)
- Kelly Nichols
- 1Animal Nutrition Group, Wageningen University and Research, PO Box 338, 6700 AH Wageningen, the Netherlands
| | - André Bannink
- 2Wageningen Livestock Research, Wageningen University and Research, PO Box 338, 6700 AH Wageningen, the Netherlands
| | - Jurgen van Baal
- 1Animal Nutrition Group, Wageningen University and Research, PO Box 338, 6700 AH Wageningen, the Netherlands
| | - Jan Dijkstra
- 1Animal Nutrition Group, Wageningen University and Research, PO Box 338, 6700 AH Wageningen, the Netherlands
| |
Collapse
|
48
|
Qi M, Wang J, Tan B, Li J, Liao S, Liu Y, Yin Y. Dietary glutamine, glutamate, and aspartate supplementation improves hepatic lipid metabolism in post-weaning piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2020; 6:124-129. [PMID: 32542191 PMCID: PMC7283369 DOI: 10.1016/j.aninu.2019.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/23/2022]
Abstract
A previous study has demonstrated that early weaning significantly suppressed hepatic glucose metabolism in piglets. Glutamate (Glu), aspartate (Asp) and glutamine (Gln) are major metabolic fuels for the small intestine and can alleviate weaning stress, and therefore might improve hepatic energy metabolism. The objective of this study was to investigate the effects of administration of Glu, Asp and Gln on the expression of hepatic genes and proteins involved in lipid metabolism in post-weaning piglets. Thirty-six weaned piglets were assigned to the following treatments: control diet (Control; basal diet + 15.90 g/kg alanine); Asp, Gln and Glu-supplemented diet (Control + AA; basal diet + 1.00 g/kg Asp + 5.00 g/kg Glu + 10.00 g/kg Gln); and the energy-restricted diet supplemented with Asp, Gln and Glu (Energy− + AA; energy deficient diet + 1.00 g/kg Asp + 5.00 g/kg Glu + 10.00 g/kg Gln). Liver samples were obtained on d 5 and 21 post-weaning. Piglets fed Energy− + AA diet had higher liver mRNA abundances of acyl-CoA oxidase 1 (ACOX1), succinate dehydrogenase (SDH), mitochondrial transcription factor A (TFAM) and sirtuin 1 (SIRT1), as well as higher protein expression of serine/threonine protein kinase 11 (LKB1), phosphor-acetyl-CoA carboxylase (P-ACC) and SIRT1 compared with piglets fed control diet (P < 0.05) on d 5 post-weaning. Control + AA diet increased liver malic enzyme 1 (ME1) and SIRT1 mRNA levels, as well as protein expression of LKB1 and P-ACC on d 5 post-weaning (P < 0.05). On d 21 post-weaning, compared to control group, Glu, Gln and Asp supplementation up-regulated the mRNA levels of ACOX1, ME1 and SIRT1 (P < 0.05). These findings indicated that dietary Glu, Gln and Asp supplementation could improve hepatic lipid metabolism to some extent, which may provide nutritional intervention for the insufficient energy intake after weaning in piglets.
Collapse
Affiliation(s)
- Ming Qi
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.,University of Chinese Academy of Sciences, Beijing 100008, China
| | - Jing Wang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Bi'e Tan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Jianjun Li
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Simeng Liao
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.,University of Chinese Academy of Sciences, Beijing 100008, China
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis 95616, CA, USA
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| |
Collapse
|
49
|
The NADP-dependent malic enzyme MaeB is a central metabolic hub controlled by the acetyl-CoA to CoASH ratio. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140462. [PMID: 32485238 DOI: 10.1016/j.bbapap.2020.140462] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 11/20/2022]
Abstract
Malic enzymes participate in key metabolic processes, the MaeB-like malic enzymes carry a catalytic inactive phosphotransacetylase domain whose function remains elusive. Here we show that acetyl-CoA directly binds and inhibits MaeB-like enzymes with a saturable profile under physiological relevant acetyl-CoA concentrations. A MaeB-like enzyme from the nitrogen-fixing bacterium Azospirillum brasilense, namely AbMaeB1, binds both acetyl-CoA and unesterified CoASH in a way that inhibition of AbMaeB1 by acetyl-CoA is relieved by increasing CoASH concentrations. Hence, AbMaeB1 senses the acetyl-CoA/CoASH ratio. We revisited E. coli MaeB regulation to determine the inhibitory constant for acetyl-CoA. Our data support that the phosphotransacetylase domain of MaeB-like enzymes senses acetyl-CoA to dictate the fate of carbon distribution at the phosphoenol-pyruvate / pyruvate / oxaloacetate metabolic node.
Collapse
|
50
|
Katagiri T, Amao Y. Trivalent metal ions promote the malic enzyme-catalyzed building of carbon–carbon bonds from CO2and pyruvate. NEW J CHEM 2020. [DOI: 10.1039/d0nj03449e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
ME is an attractive biocatalyst for building carbon–carbon bonds through carboxylation of pyruvate with CO2. The carboxylation of pyruvate with CO2was promoted by adding a trivalent metal ion. In particular, Al3+accelerates ME-catalyzed carboxylation of pyruvate with CO2.
Collapse
Affiliation(s)
| | - Yutaka Amao
- Graduate School of Science
- Osaka City University
- Osaka 558-8585
- Japan
- Research Centre of Artificial Photosynthesis (ReCAP)
| |
Collapse
|