1
|
Martin M, Kumar R, Buchkovich NJ, Norbury CC. HCMV infection downregulates GPX4 and stimulates lipid peroxidation but does not induce ferroptosis. J Virol 2025:e0185124. [PMID: 39772623 DOI: 10.1128/jvi.01851-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 11/30/2024] [Indexed: 01/11/2025] Open
Abstract
Human cytomegalovirus (HCMV) modulates numerous cellular pathways to facilitate infection, including key components in cellular iron homeostasis. Iron is essential to many cellular processes but, if present in excess, drives cell death through ferroptosis. Ferroptosis is a process that is dependent upon the accumulation of oxidatively damaged phospholipids (lipid peroxides); when these lipid peroxides accumulate in membranes, this culminates in plasma membrane rupture and eventual cell lysis. Here, we demonstrate that HCMV infection downregulates the expression of a key modulator of lipid peroxidation, glutathione peroxidase 4 (GPX4). HCMV infection also markedly increased levels of lipid peroxides within infected cells. Despite the marked downregulation of GPX4 by HCMV, further inhibition of GPX4 impaired virus replication. Interestingly, overexpression of GPX4 did not reduce the production of lipid peroxides within infected cells. In contrast, lipid peroxide levels were reduced by treatment with ferrostatin-1, a ferrous iron-dependent scavenger of alkoxyl radicals, indicating a role for iron in the production of lipid peroxides. HCMV-infected cells became less sensitive to GPX4 inhibition as infection progressed, requiring substantially higher levels of GPX4 inhibitors to induce ferroptosis compared to uninfected cells. This observed difference in sensitivity to ferroptosis upon infection correlated with a large increase in lipid production by infected cells. Therefore, the marked stimulation of lipid peroxidation by HCMV likely proceeds through a pathway that is independent of GPX4 regulation, but the ability of lipid peroxides to stimulate ferroptosis by modulating plasma membrane rupture is likely blunted by the massive increase in lipid production during HCMV infection. IMPORTANCE Human cytomegalovirus (HCMV) infection is intimately linked with countless host cell pathways that are modulated in a coordinated fashion to facilitate infection. Here, we describe HCMV-induced regulation of lipid peroxidation, a precursor of the iron-regulated cell death pathway known as ferroptosis, during human cytomegalovirus infection. These studies reveal hitherto unidentified changes in metabolism mediated by HCMV that decrease sensitivity to ferroptosis, despite increases in lipid peroxidation and transient increases in intracellular iron levels in infected cells.
Collapse
Affiliation(s)
- Madison Martin
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Rinki Kumar
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Nicholas J Buchkovich
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Christopher C Norbury
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
2
|
Zhu L, Ming H, Scatolin GN, Xiao A, Jiang Z. METTL7A improves bovine IVF embryo competence by attenuating oxidative stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.17.628915. [PMID: 39763908 PMCID: PMC11702634 DOI: 10.1101/2024.12.17.628915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
In vitro fertilization (IVF) is a widely used assisted reproductive technology to achieve a successful pregnancy. However, the acquisition of oxidative stress in embryo in vitro culture impairs its competence. Here, we demonstrated that a nuclear coding gene, methyltransferase-like protein 7A (METTL7A), improves the developmental potential of bovine embryos. We found that exogenous METTL7A modulates expression of genes involved in embryonic cell mitochondrial pathways and promotes trophectoderm development. Surprisingly, we discovered that METTL7A alleviates mitochondrial stress and DNA damage and promotes cell cycle progression during embryo cleavage. In summary, we have identified a novel mitochondria stress eliminating mechanism regulated by METTL7A that occurs during the acquisition of oxidative stress in embryo in vitro culture. This discovery lays the groundwork for the development of METTL7A as a promising therapeutic target for IVF embryo competence.
Collapse
Affiliation(s)
- Linkai Zhu
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Hao Ming
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Giovanna N. Scatolin
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Andrew Xiao
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - Zongliang Jiang
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
3
|
Luo Z, Zhou F, Jiang S, Huang J, Yang L, Yang Q, Shi J, Li E, Ma Z, Li Y. Immune and physiological responses in Penaeus monodon to ammonia-N stress: a multi-omics approach. Front Immunol 2024; 15:1510887. [PMID: 39720717 PMCID: PMC11666502 DOI: 10.3389/fimmu.2024.1510887] [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: 10/14/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024] Open
Abstract
Ammonia-N stress is a significant environmental factor that adversely affects the health and productivity of aquaculture species. This study investigates the effects of ammonia-N stress on the shrimp Penaeus monodon through a combination of biochemical, histological, transcriptomic, and metabolomic analyses. Shrimp were exposed to ammonia-N stress for 12 and 96 hours, and key markers of oxidative stress, nitrogen metabolism, immune response, and overall health were assessed. The results showed that prolonged ammonia-N exposure causes significant hepatopancreatic damage, including atrophy and deformation. Transcriptomic analysis revealed significant changes in gene expression related to apoptosis, immune response, and key metabolic pathways, with particular emphasis on the disruption of innate immune signaling and defense mechanisms. Metabolomic analysis identified disruptions in nucleotide turnover, antioxidant defenses, and fundamental metabolic processes. These findings suggest that ammonia-N stress induces a multifaceted stress response in shrimp, involving oxidative stress, immune activation, and metabolic disturbances. Understanding these immune-related and metabolic mechanisms provides valuable insights into the molecular responses of crustaceans to environmental stress, laying the foundation for assessing the ecological risk of ammonia-N and identifying potential immunological biomarkers for monitoring and mitigating its adverse effects in aquaculture systems.
Collapse
Affiliation(s)
- Zhi Luo
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Falin Zhou
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Song Jiang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Jianhua Huang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
- Shenzhen Base of South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shenzhen, China
| | - Lishi Yang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Qibin Yang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, Sanya Tropical Fisheries Research Institute, Sanya, China
- Shenzhen Base of South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shenzhen, China
| | - Jianzhi Shi
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Erchao Li
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhenhua Ma
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Yundong Li
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
- Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, Sanya Tropical Fisheries Research Institute, Sanya, China
- Shenzhen Base of South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shenzhen, China
| |
Collapse
|
4
|
Tyagi SC. Epigenetics of Homocystinuria, Hydrogen Sulfide, and Circadian Clock Ablation in Cardiovascular-Renal Disease. Curr Issues Mol Biol 2024; 46:13783-13797. [PMID: 39727952 PMCID: PMC11726923 DOI: 10.3390/cimb46120824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 12/28/2024] Open
Abstract
Morning-time heart attacks are associated with an ablation in the sleep-time dip in blood pressure, the mechanism of which is unknown. The epigenetic changes are the hallmark of sleep and circadian clock disruption and homocystinuria (HHcy). The homocystinuria causes ablation in the dip in blood pressure during sleep. Interestingly, HHcy is generated during the epigenetic gene turning off and turning on (i.e., imprinting) by methylation of the DNA promoter. The mitochondrial sulfur metabolism by 3-mercaptopyruvate sulfur transferase (3MST), ATP citrate lyase (ACYL), and epigenetic rhythmic methylation are regulated by folate 1-carbon metabolism (FOCM), i.e., the methionine (M)-SAM-SAH-Hcy, adenosine, and uric acid cycle. Epigenetic gene writer (DNMT), gene eraser (TET/FTO), and editor de-aminase (ADAR) regulate the rhythmic, i.e., reversible methylation/demethylation of H3K4, H3K9, H4K20, m6A, and m5C. The mitochondrial ATP citrate cycle and creatine kinase (CK) regulate chromatin transcription, maturation, and accessibility as well as muscle function. The transcription is regulated by methylation. The maturation and accessibility are controlled by acetylation. However, it is unclear whether a high fat dysbiotic diet (HFD) causes dysrhythmic expression of the gene writer, eraser, and editor, creating hyperuricemia and cardiac and renal dysfunction. We hypothesized that an HFD increases the gene writer (DNMT1) and editor (ADAR), decreases the eraser (TET/FTO), and increases uric acid to cause chronic diseases. This increases the levels of H3K4, H3K9, H4K20, m6A, and m5C. Interestingly, the DNMT1KO mitigates. Further, the DNMT1KO and ADAR inhibition attenuate HFD-induced NGAL/FGF23/TMPRSS2/MMP2, 9, 13, and uric acid levels and improve cardiac and renal remodeling. Although the novel role of nerve endings by the Piezo channels (i.e., the combination of ENaC, VDAC, TRPV, K+, and Mg2+ channels) in the interoception is suggested, interestingly, we and others have shown mechanisms independent of the nerve, by interoception, such as the cargo of the exosome in denervation models of heart failure. If proper and appropriate levels of these enzymes are available to covert homocysteine to hydrogen sulfide (H2S) during homocystinuria, then the H2S can potentially serve as a newer form of treatment for morning heart attacks and renal sulfur transsulfuration transport diseases.
Collapse
Affiliation(s)
- Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
5
|
Saito T, Espe M, Vikeså V, Bock C, Thomsen TH, Adam AC, Fernandes JMO, Skjaerven KH. One-carbon metabolism nutrients impact the interplay between DNA methylation and gene expression in liver, enhancing protein synthesis in Atlantic salmon. Epigenetics 2024; 19:2318517. [PMID: 38404006 PMCID: PMC10900267 DOI: 10.1080/15592294.2024.2318517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/07/2024] [Indexed: 02/27/2024] Open
Abstract
Supplementation of one-carbon (1C) metabolism micronutrients, which include B-vitamins and methionine, is essential for the healthy growth and development of Atlantic salmon (Salmo salar). However, the recent shift towards non-fish meal diets in salmon aquaculture has led to the need for reassessments of recommended micronutrient levels. Despite the importance of 1C metabolism in growth performance and various cellular regulations, the molecular mechanisms affected by these dietary alterations are less understood. To investigate the molecular effect of 1C nutrients, we analysed gene expression and DNA methylation using two types of omics data: RNA sequencing (RNA-seq) and reduced-representation bisulphite sequencing (RRBS). We collected liver samples at the end of a feeding trial that lasted 220 days through the smoltification stage, where fish were fed three different levels of four key 1C nutrients: methionine, vitamin B6, B9, and B12. Our results indicate that the dosage of 1C nutrients significantly impacts genetic and epigenetic regulations in the liver of Atlantic salmon, particularly in biological pathways related to protein synthesis. The interplay between DNA methylation and gene expression in these pathways may play an important role in the mechanisms underlying growth performance affected by 1C metabolism.
Collapse
Affiliation(s)
- Takaya Saito
- Feed and Nutrition group, Institute of Marine Research, Bergen, Norway
| | - Marit Espe
- Feed and Nutrition group, Institute of Marine Research, Bergen, Norway
| | - Vibeke Vikeså
- Skretting AI, Aquaculture Innovation, Stavanger, Norway
| | - Christoph Bock
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Anne-Catrin Adam
- Feed and Nutrition group, Institute of Marine Research, Bergen, Norway
| | | | - Kaja H Skjaerven
- Feed and Nutrition group, Institute of Marine Research, Bergen, Norway
| |
Collapse
|
6
|
Shan S, Hoffman JM. Serine metabolism in aging and age-related diseases. GeroScience 2024:10.1007/s11357-024-01444-1. [PMID: 39585647 DOI: 10.1007/s11357-024-01444-1] [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: 10/14/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024] Open
Abstract
Non-essential amino acids are often overlooked in biomedical research; however, they are crucial components of organismal metabolism. One such metabolite that is integral to physiological function is serine. Serine acts as a pivotal link connecting glycolysis with one-carbon and lipid metabolism, as well as with pyruvate and glutathione syntheses. Interestingly, increasing evidence suggests that serine metabolism may impact the aging process, and supplementation with serine may confer benefits in safeguarding against aging and age-related disorders. This review synthesizes recent insights into the regulation of serine metabolism during aging and its potential to promote healthy lifespan and mitigate a spectrum of age-related diseases.
Collapse
Affiliation(s)
- Shengshuai Shan
- Department of Biological Sciences, Augusta University, Augusta, GA, 30912, USA.
| | - Jessica M Hoffman
- Department of Biological Sciences, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
7
|
Li C, Li Y, Guo J, Wang Y, Shi X, Zhang Y, Liang N, Ma H, Yuan J, Xu J, Chen H. Abundant mRNA m 1A modification in dinoflagellates: a new layer of gene regulation. EMBO Rep 2024; 25:4655-4673. [PMID: 39223385 PMCID: PMC11549093 DOI: 10.1038/s44319-024-00234-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Dinoflagellates, a class of unicellular eukaryotic phytoplankton, exhibit minimal transcriptional regulation, representing a unique model for exploring gene expression. The biosynthesis, distribution, regulation, and function of mRNA N1-methyladenosine (m1A) remain controversial due to its limited presence in typical eukaryotic mRNA. This study provides a comprehensive map of m1A in dinoflagellate mRNA and shows that m1A, rather than N6-methyladenosine (m6A), is the most prevalent internal mRNA modification in various dinoflagellate species, with an asymmetric distribution along mature transcripts. In Amphidinium carterae, we identify 6549 m1A sites characterized by a non-tRNA T-loop-like sequence motif within the transcripts of 3196 genes, many of which are involved in regulating carbon and nitrogen metabolism. Enriched within 3'UTRs, dinoflagellate mRNA m1A levels negatively correlate with translation efficiency. Nitrogen depletion further decreases mRNA m1A levels. Our data suggest that distinctive patterns of m1A modification might influence the expression of metabolism-related genes through translational control.
Collapse
Affiliation(s)
- Chongping Li
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong & Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China
| | - Ying Li
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong & Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China
- Shenzhen People's Hospital, 3046 Shennan E Rd, Shenzhen, 518020, China
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Jia Guo
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China
- NHC Key Laboratory of Birth Defects Prevention, Zhengzhou, 450000, China
| | - Yuci Wang
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong & Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Xiaoyan Shi
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong & Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China
| | - Yangyi Zhang
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong & Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China
| | - Nan Liang
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong & Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China
| | - Honghui Ma
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Institutes of Biomedical Sciences, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Key Laboratory of Medical Epigenetics and Metabolism, Fudan University, Shanghai, 200000, China
| | - Jie Yuan
- Shenzhen People's Hospital, 3046 Shennan E Rd, Shenzhen, 518020, China.
| | - Jiawei Xu
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China.
- NHC Key Laboratory of Birth Defects Prevention, Zhengzhou, 450000, China.
| | - Hao Chen
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong & Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China.
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China.
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, 518000, China.
- NHC Key Laboratory of Birth Defects Prevention, Zhengzhou, 450000, China.
| |
Collapse
|
8
|
Kriström K, Häggström J, Fascetti AJ, Ström L, Dirven M, Yu J, Essén TS, Tidholm A, Pion PD, Ljungvall I. The association between taurine concentrations and dog characteristics, clinical variables, and diet in English cocker spaniels: The Canine taURinE (CURE) project. J Vet Intern Med 2024; 38:2620-2632. [PMID: 39136304 PMCID: PMC11423459 DOI: 10.1111/jvim.17150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 07/16/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Occurrence of low blood taurine concentrations (B-TauC) and predisposing factors to taurine deficiency in English Cocker Spaniels (ECS) are incompletely understood. OBJECTIVES Investigate the occurrence of low B-TauC in a Swedish population of ECS and evaluate the association between B-TauC and dog characteristics, clinical variables, and diet composition. ANIMALS One-hundred eighty privately owned ECS. METHODS Dogs were prospectively recruited and underwent physical examination, blood analyses, and echocardiographic and ophthalmic examinations. Dogs with clinical signs of congestive heart failure (CHF) also underwent thoracic radiography. Taurine concentrations were analyzed in plasma (EDTA and heparin) and whole blood. Diets consumed by the dogs at the time of the examination were analyzed for dietary taurine- (D-TauC), cysteine- (D-CysC), and methionine concentrations (D-MetC). RESULTS Fifty-three of 180 dogs (29%) had low B-TauC, of which 13 (25%) dogs had clinical and radiographic signs of CHF, increased echocardiographic left ventricular (LV) dimensions and volumes, and impaired LV systolic function. Five (9%) dogs with low B-TauC had retinal abnormalities. Dietary MetC, dietary animal protein source (red/white meat), and age were associated with B-TauC in the final multivariable regression model (P < .001, R2 adj = .39). CONCLUSIONS AND CLINICAL IMPORTANCE Low B-TauC suggests that taurine deficiency may play a role in the development of myocardial failure and CHF in ECS. Low D-MetC and diets with red meat as the animal protein source were associated with low B-TauC. Dogs with B-TauC below the normal reference range were older than dogs with normal concentrations.
Collapse
Affiliation(s)
- Karin Kriström
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
- Evidensia Södra Animal Hospital, Huddinge, Sweden
| | - Jens Häggström
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Andrea J Fascetti
- Department of Molecular Biosciences, School of Veterinary Medicine University of California Davis, Davis, California, USA
| | - Lena Ström
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Mark Dirven
- Evidensia Södra Animal Hospital, Huddinge, Sweden
| | - Joshua Yu
- Department of Molecular Biosciences, School of Veterinary Medicine University of California Davis, Davis, California, USA
| | | | - Anna Tidholm
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
- Anicura Albano Animal Hospital, Danderyd, Sweden
| | - Paul D Pion
- Veterinary Information Network, Davis, California, USA
| | - Ingrid Ljungvall
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
9
|
Yao H, Jiang W, Liao X, Wang D, Zhu H. Regulatory mechanisms of amino acids in ferroptosis. Life Sci 2024; 351:122803. [PMID: 38857653 DOI: 10.1016/j.lfs.2024.122803] [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: 03/12/2024] [Revised: 05/19/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Ferroptosis, an iron-dependent non-apoptotic regulated cell death process, is associated with the pathogenesis of various diseases. Amino acids, which are indispensable substrates of vital activities, significantly regulate ferroptosis. Amino acid metabolism is involved in maintaining iron and lipid homeostasis and redox balance. The regulatory effects of amino acids on ferroptosis are complex. An amino acid may exert contrasting effects on ferroptosis depending on the context. This review systematically and comprehensively summarized the distinct roles of amino acids in regulating ferroptosis and highlighted the emerging opportunities to develop clinical therapeutic strategies targeting amino acid-mediated ferroptosis.
Collapse
Affiliation(s)
- Heying Yao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Wei Jiang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Xiang Liao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Dongqing Wang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
10
|
Pasqualette L, Fidalgo TKDS, Freitas-Fernandes LB, Souza GGL, Imbiriba LA, Lobo LA, Volchan E, Domingues RMCP, Valente AP, Miranda KR. Alterations in Vagal Tone Are Associated with Changes in the Gut Microbiota of Adults with Anxiety and Depression Symptoms: Analysis of Fecal Metabolite Profiles. Metabolites 2024; 14:450. [PMID: 39195546 DOI: 10.3390/metabo14080450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
Accumulating evidence suggests that interactions between the brain and gut microbiota significantly impact brain function and mental health. In the present study, we aimed to investigate whether young, healthy adults without psychiatric diagnoses exhibit differences in metabolic stool and microbiota profiles based on depression/anxiety scores and heart rate variability (HRV) parameters. Untargeted nuclear magnetic resonance-based metabolomics was used to identify fecal metabolic profiles. Results were subjected to multivariate analysis through principal component analysis (PCA) and partial least squares discriminant analysis (PLS-DA), and the metabolites were identified through VIP score. Metabolites separating asymptomatic and symptomatic groups were acetate, valine, and glutamate, followed by sugar regions, glutamine, acetone, valerate, and acetoacetate. The main metabolites identified in high vagal tone (HVT) and low vagal tone (LVT) groups were acetate, valerate, and glutamate, followed by propionate and butyrate. In addition to the metabolites identified by the PLS-DA test, significant differences in aspartate, sarcosine, malate, and methionine were observed between the groups. Levels of acetoacetate were higher in both symptomatic and LVT groups. Valerate levels were significantly increased in the symptomatic group, while isovalerate, propionate, glutamate, and acetone levels were significantly increased in the LVT group. Furthermore, distinct abundance between groups was only confirmed for the Firmicutes phylum. Differences between participants with high and low vagal tone suggest that certain metabolites are involved in communication between the vagus nerve and the brain.
Collapse
Affiliation(s)
- Laura Pasqualette
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Developmental and Educational Psychology, University of Bremen, 28359 Bremen, Germany
| | - Tatiana Kelly da Silva Fidalgo
- Pediatric Dentistry, Department of Preventive and Community Dentistry, State University of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Liana Bastos Freitas-Fernandes
- National Centre of Nuclear Magnetic Resonance/CENABIO, Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Gabriela Guerra Leal Souza
- Laboratory of Psychophysiology, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil
| | - Luís Aureliano Imbiriba
- School of Physical Education and Sports, Federal University of Rio de Janeiro, Rio de Janeiro 21941-599, Brazil
| | - Leandro Araujo Lobo
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Eliane Volchan
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | | | - Ana Paula Valente
- National Centre of Nuclear Magnetic Resonance/CENABIO, Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Karla Rodrigues Miranda
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
11
|
Prigent M, Jean-Jacques H, Naquin D, Chédin S, Cuif MH, Legouis R, Kuras L. Sulfur starvation-induced autophagy in Saccharomyces cerevisiae involves SAM-dependent signaling and transcription activator Met4. Nat Commun 2024; 15:6927. [PMID: 39138175 PMCID: PMC11322535 DOI: 10.1038/s41467-024-51309-6] [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: 05/03/2022] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
Autophagy is a key lysosomal degradative mechanism allowing a prosurvival response to stresses, especially nutrient starvation. Here we investigate the mechanism of autophagy induction in response to sulfur starvation in Saccharomyces cerevisiae. We found that sulfur deprivation leads to rapid and widespread transcriptional induction of autophagy-related (ATG) genes in ways not seen under nitrogen starvation. This distinctive response depends mainly on the transcription activator of sulfur metabolism Met4. Consistently, Met4 is essential for autophagy under sulfur starvation. Depletion of either cysteine, methionine or SAM induces autophagy flux. However, only SAM depletion can trigger strong transcriptional induction of ATG genes and a fully functional autophagic response. Furthermore, combined inactivation of Met4 and Atg1 causes a dramatic decrease in cell survival under sulfur starvation, highlighting the interplay between sulfur metabolism and autophagy to maintain cell viability. Thus, we describe a pathway of sulfur starvation-induced autophagy depending on Met4 and involving SAM as signaling sulfur metabolite.
Collapse
Affiliation(s)
- Magali Prigent
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
- INSERM U1280, 91198, Gif-sur-Yvette, France
| | - Hélène Jean-Jacques
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Delphine Naquin
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Stéphane Chédin
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Marie-Hélène Cuif
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
- INSERM U1280, 91198, Gif-sur-Yvette, France
| | - Renaud Legouis
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
- INSERM U1280, 91198, Gif-sur-Yvette, France
| | - Laurent Kuras
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France.
| |
Collapse
|
12
|
Fang N, Liu B, Pan Q, Gong T, Zhan M, Zhao J, Wang Q, Tang Y, Li Y, He J, Xiang T, Sun F, Lu L, Xia J. SMG5 Inhibition Restrains Hepatocellular Carcinoma Growth and Enhances Sorafenib Sensitivity. Mol Cancer Ther 2024; 23:1188-1200. [PMID: 38647536 DOI: 10.1158/1535-7163.mct-23-0729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/25/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
Hepatocellular carcinoma (HCC) has a pathogenesis that remains elusive with restricted therapeutic strategies and efficacy. This study aimed to investigate the role of SMG5, a crucial component in nonsense-mediated mRNA decay (NMD) that degrades mRNA containing a premature termination codon, in HCC pathogenesis and therapeutic resistance. We demonstrated an elevated expression of SMG5 in HCC and scrutinized its potential as a therapeutic target. Our findings revealed that SMG5 knockdown not only inhibited the migration, invasion, and proliferation of HCC cells but also influenced sorafenib resistance. Differential gene expression analysis between the control and SMG5 knockdown groups showed an upregulation of methionine adenosyltransferase 1A in the latter. High expression of methionine adenosyltransferase 1A, a catalyst for S-adenosylmethionine (SAM) production, as suggested by The Cancer Genome Atlas data, was indicative of a better prognosis for HCC. Further, an ELISA showed a higher concentration of SAM in SMG5 knockdown cell supernatants. Furthermore, we found that exogenous SAM supplementation enhanced the sensitivity of HCC cells to sorafenib alongside changes in the expression of Bax and Bcl-2, apoptosis-related proteins. Our findings underscore the important role of SMG5 in HCC development and its involvement in sorafenib resistance, highlighting it as a potential target for HCC treatment.
Collapse
Affiliation(s)
- Nan Fang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, P. R. China
| | - Bing Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, P. R. China
| | - Qiuzhong Pan
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Tingting Gong
- Department of Ultrasound, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, P. R. China
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, P. R. China
| | - Jingjing Zhao
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Qijing Wang
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yan Tang
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yongqiang Li
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jia He
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Tong Xiang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Fengze Sun
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, P. R. China
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, P. R. China
| | - Jianchuan Xia
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| |
Collapse
|
13
|
Fogal V, Michopoulos F, Jarnuczak AF, Hamza GM, Harlfinger S, Davey P, Hulme H, Atkinson SJ, Gabrowski P, Cheung T, Grondine M, Hoover C, Rose J, Bray C, Foster AJ, Askin S, Majumder MM, Fitzpatrick P, Miele E, Macdonald R, Keun HC, Coen M. Mechanistic safety assessment via multi-omic characterisation of systemic pathway perturbations following in vivo MAT2A inhibition. Arch Toxicol 2024; 98:2589-2603. [PMID: 38755480 PMCID: PMC11272821 DOI: 10.1007/s00204-024-03771-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
The tumour suppressor p16/CDKN2A and the metabolic gene, methyl-thio-adenosine phosphorylase (MTAP), are frequently co-deleted in some of the most aggressive and currently untreatable cancers. Cells with MTAP deletion are vulnerable to inhibition of the metabolic enzyme, methionine-adenosyl transferase 2A (MAT2A), and the protein arginine methyl transferase (PRMT5). This synthetic lethality has paved the way for the rapid development of drugs targeting the MAT2A/PRMT5 axis. MAT2A and its liver- and pancreas-specific isoform, MAT1A, generate the universal methyl donor S-adenosylmethionine (SAM) from ATP and methionine. Given the pleiotropic role SAM plays in methylation of diverse substrates, characterising the extent of SAM depletion and downstream perturbations following MAT2A/MAT1A inhibition (MATi) is critical for safety assessment. We have assessed in vivo target engagement and the resultant systemic phenotype using multi-omic tools to characterise response to a MAT2A inhibitor (AZ'9567). We observed significant SAM depletion and extensive methionine accumulation in the plasma, liver, brain and heart of treated rats, providing the first assessment of both global SAM depletion and evidence of hepatic MAT1A target engagement. An integrative analysis of multi-omic data from liver tissue identified broad perturbations in pathways covering one-carbon metabolism, trans-sulfuration and lipid metabolism. We infer that these pathway-wide perturbations represent adaptive responses to SAM depletion and confer a risk of oxidative stress, hepatic steatosis and an associated disturbance in plasma and cellular lipid homeostasis. The alterations also explain the dramatic increase in plasma and tissue methionine, which could be used as a safety and PD biomarker going forward to the clinic.
Collapse
Affiliation(s)
- Valentina Fogal
- Oncology Safety, Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Cancer Metabolism & Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Filippos Michopoulos
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Andrew F Jarnuczak
- Data Sciences & Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Ghaith M Hamza
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, R&D Boston, Waltham, USA
| | | | - Paul Davey
- Chemistry, Oncology R&D AstraZeneca, Cambridge, UK
| | - Heather Hulme
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | | | - Piotr Gabrowski
- Biological Insights Knowledge Graph, R&D IT, AstraZeneca, Barcelona, Spain
| | - Tony Cheung
- Oncology R&D, AstraZeneca, R&D Boston, Waltham, USA
| | | | - Clare Hoover
- Oncology Safety Pathology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, R&D Boston, Waltham, USA
| | - Jonathan Rose
- Animal Science & Technologies, R&D, AstraZeneca, Cambridge, UK
| | - Chandler Bray
- Cancer Metabolism & Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Alison J Foster
- Regulatory Toxicology and Safety Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Sean Askin
- Advanced Drug Delivery, Pharmaceutical Sci, R&D, AstraZeneca, Cambridge, UK
| | - Muntasir Mamun Majumder
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Paul Fitzpatrick
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Eric Miele
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, R&D Boston, Waltham, USA
| | - Ruth Macdonald
- Animal Science & Technologies, R&D, AstraZeneca, Cambridge, UK
| | - Hector C Keun
- Cancer Metabolism & Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Muireann Coen
- Oncology Safety, Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
14
|
Das BK, Ganguly S, Bayen S, Talukder AK, Ray A, Das Gupta S, Kumari K. Amino Acid Composition of Thirty Food Fishes of the Ganga Riverine Environment for Addressing Amino Acid Requirement through Fish Supplementation. Foods 2024; 13:2124. [PMID: 38998630 PMCID: PMC11241810 DOI: 10.3390/foods13132124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/14/2024] Open
Abstract
Amino acids are significant biomolecules that govern the major metabolic processes and act as precursors for macromolecules such as proteins that are crucial to life. Fish is an integral component of human nutrition and a dietary source of high-quality animal proteins and amino acids. In this context, the crude protein and amino acid compositions of food fish from different landing stations of the Ganga river have been determined. The Kjeldahl method was utilized to determine the crude protein content and the amino acids were analyzed using high-performance liquid chromatography (HPLC); data on 30 food fish were assessed. The study showed that among the fish studied, Eleotris fusca, Macrobrachium malcomsonii, and Mystus cavasius were rich in most of the amino acids important for human nutrition, such as glycine, glutamic acid, cysteine, threonine, phenylalanine, methionine, lysine, leucine, isoleucine, histidine, and valine. Further, it was observed that the daily consumption of these fish (approximately 50 g) can fulfil the daily requirement of these individual amino acids for an adult human with a body weight of 60 kg. Therefore, the amino acid composition analyzed in the present study could be utilized for recommendation by clinicians according to the requirement for specific amino acids, and fish can be prescribed as a natural supplement against the amino acid requirement.
Collapse
Affiliation(s)
- Basanta Kumar Das
- ICAR-Central Inland Fisheries Research Institute, Kolkata 700120, India
| | | | | | | | | | | | | |
Collapse
|
15
|
Cotton K, Ayers E, Jin Y, Beauchet O, Derby CA, Lipton RB, Katz M, Galery K, Gaudreau P, Verghese J. Elevated Blood Homocysteine Increases the Risk of Incident Motoric Cognitive Risk Syndrome: A Two-Cohort Study. J Gerontol A Biol Sci Med Sci 2024; 79:glae114. [PMID: 38671552 PMCID: PMC11157967 DOI: 10.1093/gerona/glae114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Motoric Cognitive Risk (MCR) syndrome, a predementia syndrome characterized by cognitive complaints and slow gait, may have an underlying vascular etiology. Elevated blood levels of homocysteine, a known vascular risk factor, have been linked to physical and cognitive decline in older adults, though the relationship with MCR is unknown. We aimed to identify the association between homocysteine and MCR risk. METHODS We examined the association between baseline homocysteine levels and incident MCR using Cox proportional hazard models in 1826 community-dwelling older adults (55% women) from 2 cohorts (Einstein Aging Study [EAS] and Quebec Longitudinal Study on Nutrition and Successful Aging [NuAge]). We calculated hazard ratios (HR) with 95% confidence intervals (CI), for each cohort as well as stratified by sex and vascular disease/risk factors. RESULTS Median follow-up time was 2.2 years in EAS and 3.0 years in NuAge. Individuals with elevated baseline homocysteine levels (>14 µmol/L) had a significantly higher risk of incident MCR compared to those with normal levels in NuAge (HR 1.41, 95% CI: 1.01-1.97, p = .04), after adjusting for covariates. Our exploratory stratified analyses found that these associations were significant only in men with vascular disease/risk factors. CONCLUSIONS Higher blood homocysteine levels are associated with an increased risk of developing MCR in older adults, particularly in men with vascular disease or vascular risk factors.
Collapse
Affiliation(s)
- Kelly Cotton
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Emmeline Ayers
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ying Jin
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Olivier Beauchet
- Research Centre of the Geriatric University Institute of Montreal, Montreal, Quebec, Canada
- Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Carol A Derby
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Richard B Lipton
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Mindy Katz
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kevin Galery
- Research Centre of the Geriatric University Institute of Montreal, Montreal, Quebec, Canada
| | - Pierrette Gaudreau
- Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Centre Hospitalier de l’Université de Montréal Research Center, Montreal, Quebec, Canada
| | - Joe Verghese
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
16
|
Caracausi M, Ramacieri G, Catapano F, Cicilloni M, Lajin B, Pelleri MC, Piovesan A, Vitale L, Locatelli C, Pirazzoli GL, Strippoli P, Antonaros F, Vione B. The functional roles of S-adenosyl-methionine and S-adenosyl-homocysteine and their involvement in trisomy 21. Biofactors 2024; 50:709-724. [PMID: 38353465 DOI: 10.1002/biof.2044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/03/2024] [Indexed: 08/09/2024]
Abstract
The one-carbon metabolism pathway is involved in critical human cellular functions such as cell proliferation, mitochondrial respiration, and epigenetic regulation. In the homocysteine-methionine cycle S-adenosyl-methionine (SAM) and S-adenosyl-homocysteine (SAH) are synthetized, and their levels are finely regulated to ensure proper functioning of key enzymes which control cellular growth and differentiation. Here we review the main biological mechanisms involving SAM and SAH and the known related human diseases. It was recently demonstrated that SAM and SAH levels are altered in plasma of subjects with trisomy 21 (T21) but how this metabolic dysregulation influences the clinical manifestation of T21 phenotype has not been previously described. This review aims at providing an overview of the biological mechanisms which are altered in response to changes in the levels of SAM and SAH observed in DS.
Collapse
Affiliation(s)
- Maria Caracausi
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Giuseppe Ramacieri
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Speciality School of Child Neuropsychiatry-Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Francesca Catapano
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Michela Cicilloni
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Bassam Lajin
- Institute of Chemistry, ChromICP, University of Graz, Graz, Austria
| | - Maria Chiara Pelleri
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Allison Piovesan
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Lorenza Vitale
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Chiara Locatelli
- Neonatology Unit, St. Orsola-Malpighi Polyclinic, Bologna, Italy
| | | | - Pierluigi Strippoli
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Francesca Antonaros
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Beatrice Vione
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| |
Collapse
|
17
|
Majtan T, Olsen T, Sokolova J, Krijt J, Křížková M, Ida T, Ditrói T, Hansikova H, Vit O, Petrak J, Kuchař L, Kruger WD, Nagy P, Akaike T, Kožich V. Deciphering pathophysiological mechanisms underlying cystathionine beta-synthase-deficient homocystinuria using targeted metabolomics, liver proteomics, sphingolipidomics and analysis of mitochondrial function. Redox Biol 2024; 73:103222. [PMID: 38843767 PMCID: PMC11190558 DOI: 10.1016/j.redox.2024.103222] [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: 02/29/2024] [Revised: 05/17/2024] [Accepted: 06/02/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Cystathionine β-synthase (CBS)-deficient homocystinuria (HCU) is an inherited disorder of sulfur amino acid metabolism with varying severity and organ complications, and a limited knowledge about underlying pathophysiological processes. Here we aimed at getting an in-depth insight into disease mechanisms using a transgenic mouse model of HCU (I278T). METHODS We assessed metabolic, proteomic and sphingolipidomic changes, and mitochondrial function in tissues and body fluids of I278T mice and WT controls. Furthermore, we evaluated the efficacy of methionine-restricted diet (MRD) in I278T mice. RESULTS In WT mice, we observed a distinct tissue/body fluid compartmentalization of metabolites with up to six-orders of magnitude differences in concentrations among various organs. The I278T mice exhibited the anticipated metabolic imbalance with signs of an increased production of hydrogen sulfide and disturbed persulfidation of free aminothiols. HCU resulted in a significant dysregulation of liver proteome affecting biological oxidations, conjugation of compounds, and metabolism of amino acids, vitamins, cofactors and lipids. Liver sphingolipidomics indicated upregulation of the pro-proliferative sphingosine-1-phosphate signaling pathway. Liver mitochondrial function of HCU mice did not seem to be impaired compared to controls. MRD in I278T mice improved metabolic balance in all tissues and substantially reduced dysregulation of liver proteome. CONCLUSION The study highlights distinct tissue compartmentalization of sulfur-related metabolites in normal mice, extensive metabolome, proteome and sphingolipidome disruptions in I278T mice, and the efficacy of MRD to alleviate some of the HCU-related biochemical abnormalities.
Collapse
Affiliation(s)
- Tomas Majtan
- Department of Pharmacology, University of Fribourg, Faculty of Science and Medicine, Fribourg, 1700, Switzerland.
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jitka Sokolova
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in Prague, Prague, 12808, Czech Republic
| | - Jakub Krijt
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in Prague, Prague, 12808, Czech Republic
| | - Michaela Křížková
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in Prague, Prague, 12808, Czech Republic
| | - Tomoaki Ida
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Tamás Ditrói
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Hana Hansikova
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in Prague, Prague, 12808, Czech Republic
| | - Ondrej Vit
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
| | - Jiri Petrak
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
| | - Ladislav Kuchař
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in Prague, Prague, 12808, Czech Republic
| | - Warren D Kruger
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology Research Group, University of Veterinary Medicine, 1078, Budapest, Hungary; Chemistry Institute, University of Debrecen, 4012, Debrecen, Hungary
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Viktor Kožich
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic.
| |
Collapse
|
18
|
Nam C, Li LY, Yang Q, Ziman B, Zhao H, Hu B, Collet C, Jing P, Lei Q, Xu LY, Li EM, Koeffler HP, Sinha UK, Lin DC. A druggable cascade links methionine metabolism to epigenomic reprogramming in squamous cell carcinoma. Proc Natl Acad Sci U S A 2024; 121:e2320835121. [PMID: 38900797 PMCID: PMC11214090 DOI: 10.1073/pnas.2320835121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Upper aerodigestive squamous cell carcinoma (UASCC) is a common and aggressive malignancy with few effective therapeutic options. Here, we investigate amino acid metabolism in this cancer, surprisingly noting that UASCC exhibits the highest methionine level across all human cancers, driven by its transporter LAT1. We show that LAT1 is also expressed at the highest level in UASCC, transcriptionally activated by UASCC-specific promoter and enhancers, which are directly coregulated by SCC master regulators TP63/KLF5/SREBF1. Unexpectedly, unbiased bioinformatic screen identifies EZH2 as the most significant target downstream of the LAT1-methionine pathway, directly linking methionine metabolism to epigenomic reprogramming. Importantly, this cascade is indispensable for the survival and proliferation of UASCC patient-derived tumor organoids. In addition, LAT1 expression is closely associated with cellular sensitivity to inhibition of the LAT1-methionine-EZH2 axis. Notably, this unique LAT1-methionine-EZH2 cascade can be targeted effectively by either pharmacological approaches or dietary intervention in vivo. In summary, this work maps a unique mechanistic cross talk between epigenomic reprogramming with methionine metabolism, establishes its biological significance in the biology of UASCC, and identifies a unique tumor-specific vulnerability which can be exploited both pharmacologically and dietarily.
Collapse
Affiliation(s)
- Chehyun Nam
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA90033
| | - Li-Yan Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou515041, Guangdong, China
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Qian Yang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Benjamin Ziman
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA90033
| | - Hua Zhao
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA90033
| | - Boyan Hu
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA90033
| | - Casey Collet
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA90033
| | - Pei Jing
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou515041, Guangdong, China
| | - Qifang Lei
- Department of Urology, South China Hospital of Shenzhen University, Shenzhen, Guangdong518116, China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou515041, Guangdong, China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou515041, Guangdong, China
| | | | - Uttam K. Sinha
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - De-Chen Lin
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA90033
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA90048
| |
Collapse
|
19
|
Golabi M, Kazemi D, Chadeganipour AS, Fouladseresht H, Sullman MJM, Ghezelbash B, Dastgerdi AY, Eskandari N. The Role of Cobalamin in Multiple Sclerosis: An Update. Inflammation 2024:10.1007/s10753-024-02075-6. [PMID: 38902541 DOI: 10.1007/s10753-024-02075-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024]
Abstract
Multiple sclerosis (MS) is a neurodegenerative condition that results in axonal and permanent damage to the central nervous system, necessitating healing owing to autoimmune reactions and persistent neuroinflammation. Antioxidant and anti-inflammatory drugs are essential for the management of oxidative stress and neuroinflammation. Additionally, multivitamin supplementation, particularly vitamin B12 (cobalamin), may be beneficial for neuronal protection. Although there is no documented connection between vitamin B12 deficiency and MS, researchers have explored its potential as a metabolic cause. This review highlights the therapeutic benefits of cobalamin (Cbl) in patients with MS.
Collapse
Affiliation(s)
- Marjan Golabi
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Danial Kazemi
- Student Research Committee, Isfahan University of Medical Science, Isfahan, Iran
| | | | - Hamed Fouladseresht
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mark J M Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Behrooz Ghezelbash
- Laboratory Hematology and Blood Banking, School of Allied Medical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ava Yeganegi Dastgerdi
- Department of Cell and Molecular Biology, Falavarjan Branch, Islamic Azad University of Science, Isfahan, Iran
| | - Nahid Eskandari
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
20
|
Goicoechea L, Torres S, Fàbrega L, Barrios M, Núñez S, Casas J, Fabrias G, García-Ruiz C, Fernández-Checa JC. S-Adenosyl-l-methionine restores brain mitochondrial membrane fluidity and GSH content improving Niemann-Pick type C disease. Redox Biol 2024; 72:103150. [PMID: 38599016 PMCID: PMC11022094 DOI: 10.1016/j.redox.2024.103150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/15/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
Niemann-Pick type C (NPC) disease is a lysosomal storage disorder characterized by impaired motor coordination due to neurological defects and cerebellar dysfunction caused by the accumulation of cholesterol in endolysosomes. Besides the increase in lysosomal cholesterol, mitochondria are also enriched in cholesterol, which leads to decreased membrane fluidity, impaired mitochondrial function and loss of GSH, and has been shown to contribute to the progression of NPC disease. S-Adenosyl-l-methionine (SAM) regulates membrane physical properties through the generation of phosphatidylcholine (PC) from phosphatidylethanolamine (PE) methylation and functions as a GSH precursor by providing cysteine in the transsulfuration pathway. However, the role of SAM in NPC disease has not been investigated. Here we report that Npc1-/- mice exhibit decreased brain SAM levels but unchanged S-adenosyl-l-homocysteine content and lower expression of Mat2a. Brain mitochondria from Npc1-/- mice display decreased mitochondrial GSH levels and liquid chromatography-high resolution mass spectrometry analysis reveal a lower PC/PE ratio in mitochondria, contributing to increased mitochondrial membrane order. In vivo treatment of Npc1-/- mice with SAM restores SAM levels in mitochondria, resulting in increased PC/PE ratio, mitochondrial membrane fluidity and subsequent replenishment of mitochondrial GSH levels. In vivo SAM treatment improves the decline of locomotor activity, increases Purkinje cell survival in the cerebellum and extends the average and maximal life spam of Npc1-/- mice. These findings identify SAM as a potential therapeutic approach for the treatment of NPC disease.
Collapse
Affiliation(s)
- Leire Goicoechea
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Sandra Torres
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Laura Fàbrega
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Mónica Barrios
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Susana Núñez
- Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Departament de Química Orgànica Biològica, Institut D'Investigacions Químiques I Ambientals de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Gemma Fabrias
- Research Unit on BioActive Molecules (RUBAM), Departament de Química Orgànica Biològica, Institut D'Investigacions Químiques I Ambientals de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Carmen García-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain; Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| | - José C Fernández-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain; Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
21
|
Xue J, Ye C. The role of lipoylation in mitochondrial adaptation to methionine restriction. Bioessays 2024; 46:e2300218. [PMID: 38616332 DOI: 10.1002/bies.202300218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024]
Abstract
Dietary methionine restriction (MR) is associated with a spectrum of health-promoting benefits. Being conducive to prevention of chronic diseases and extension of life span, MR can activate integrated responses at metabolic, transcriptional, and physiological levels. However, how the mitochondria of MR influence metabolic phenotypes remains elusive. Here, we provide a summary of cellular functions of methionine metabolism and an overview of the current understanding of effector mechanisms of MR, with a focus on the aspect of mitochondria-mediated responses. We propose that mitochondria can sense and respond to MR through a modulatory role of lipoylation, a mitochondrial protein modification sensitized by MR.
Collapse
Affiliation(s)
- Jingyuan Xue
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Cunqi Ye
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Hainan Institute, Zhejiang University, Sanya, China
- National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, China
| |
Collapse
|
22
|
Kim HY, Moon JO, Kim SW. Development and application of a multi-step porcine in vitro system to evaluate feedstuffs and feed additives for their efficacy in nutrient digestion, digesta characteristics, and intestinal immune responses. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:265-282. [PMID: 38800740 PMCID: PMC11127235 DOI: 10.1016/j.aninu.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 05/29/2024]
Abstract
In vitro model provides alternatives to the use of live animals in research. In pig nutrition, there has been a tremendous increase in in vivo research over the decades. Proper utilization of in vitro models could provide a screening tool to reduce the needs of in vivo studies, research duration, cost, and the use of animals and feeds. This study aimed to develop a multi-step porcine in vitro system to simulate nutrient digestion and intestinal epithelial immune responses affected by feedstuffs and feed additives. Seven feedstuffs (corn, corn distillers dried grains with solubles [corn DDGS], barley, wheat, soybean meal, soy protein concentrates, and Corynebacterium glutamicum cell mass [CGCM]), feed enzymes (xylanase and phytase), and supplemental amino acids (arginine, methionine, and tryptophan), were used in this in vitro evaluation for their efficacy on digestibility, digesta characteristics, and intestinal health compared with the results from previously published in vivo studies. All in vitro evaluations were triplicated. Data were analyzed using Mixed procedure of SAS9.4. Evaluations included (1) nutrient digestibility of feedstuffs, (2) the effects of feed enzymes, xylanase and phytase, on digestibility of feedstuffs and specific substrates, and (3) the effects of amino acids, arginine, tryptophan, and methionine, on anti-inflammatory, anti-oxidative, and anti-heat stress statuses showing their effects (P < 0.05) on the measured items. Differences in dry matter and crude protein digestibility among the feedstuffs as well as effects of xylanase and phytase were detected (P < 0.05), including xylo-oligosaccharide profiles and phosphorus release from phytate. Supplementation of arginine, tryptophan, and methionine modulated (P < 0.05) cellular inflammatory and oxidative stress responses. The use of this in vitro model allowed the use of 3 experimental replications providing sufficient statistical power at P < 0.05. This indicates in vitro models can have increased precision and consistency compared with in vivo animal studies.
Collapse
Affiliation(s)
- Hee Yeon Kim
- Application Center, CJ Blossom Park, Suwon, South Korea
| | - Jun-Ok Moon
- Application Center, CJ Blossom Park, Suwon, South Korea
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
23
|
Sternberg Z. Neurodegenerative Etiology of Aromatic L-Amino Acid Decarboxylase Deficiency: a Novel Concept for Expanding Treatment Strategies. Mol Neurobiol 2024; 61:2996-3018. [PMID: 37953352 DOI: 10.1007/s12035-023-03684-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 09/29/2023] [Indexed: 11/14/2023]
Abstract
Aromatic l-amino acid decarboxylase deficiency (AADC-DY) is caused by one or more mutations in the DDC gene, resulting in the deficit in catecholamines and serotonin neurotransmitters. The disease has limited therapeutic options with relatively poor clinical outcomes. Accumulated evidence suggests the involvement of neurodegenerative mechanisms in the etiology of AADC-DY. In the absence of neurotransmitters' neuroprotective effects, the accumulation and the chronic presence of several neurotoxic metabolites including 4-dihydroxy-L-phenylalanine, 3-methyldopa, and homocysteine, in the brain of subjects with AADC-DY, promote oxidative stress and reduce the cellular antioxidant and methylation capacities, leading to glial activation and mitochondrial dysfunction, culminating to neuronal injury and death. These pathophysiological processes have the potential to hinder the clinical efficacy of treatments aimed at increasing neurotransmitters' synthesis and or function. This review describes in detail the mechanisms involved in AADC-DY neurodegenerative etiology, highlighting the close similarities with those involved in other neurodegenerative diseases. We then offer novel strategies for the treatment of the disease with the objective to either reduce the level of the metabolites or counteract their prooxidant and neurotoxic effects. These treatment modalities used singly or in combination, early in the course of the disease, will minimize neuronal injury, preserving the functional integrity of neurons, hence improving the clinical outcomes of both conventional and unconventional interventions in AADC-DY. These modalities may not be limited to AADC-DY but also to other metabolic disorders where a specific mutation leads to the accumulation of prooxidant and neurotoxic metabolites.
Collapse
Affiliation(s)
- Zohi Sternberg
- Jacobs School of Medicine and Biomedical Sciences, Buffalo Medical Center, Buffalo, NY, 14203, USA.
| |
Collapse
|
24
|
Hruby Weston A, Teixeira IAMA, Yoder PS, Pilonero T, Hanigan MD. Valine and nonessential amino acids affect bidirectional transport rates of leucine and isoleucine in bovine mammary epithelial cells. J Dairy Sci 2024; 107:2026-2046. [PMID: 37863296 DOI: 10.3168/jds.2023-23447] [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: 03/02/2023] [Accepted: 09/27/2023] [Indexed: 10/22/2023]
Abstract
A more complete understanding of the mechanisms controlling AA transport in mammary glands of dairy cattle will help identify solutions to increase nitrogen feeding efficiency on farms. It was hypothesized that Ala, Gln, and Gly (NEAAG), which are actively transported into cells and exchanged for all branched-chain AA (BCAA), may stimulate transport of BCAA, and that Val may antagonize transport of the other BCAA due to transporter competition. Thus, we evaluated the effects of varying concentrations of NEAAG and Val on transport and metabolism of the BCAA Ala, Met, Phe, and Thr by bovine mammary epithelial cells. Primary cultures of bovine mammary epithelial cells were assigned to treatments of low (70% of mean in vivo plasma concentrations of lactating dairy cows) and high (200%) concentrations of Val and NEAAG (LVal and LNEAAG, HVal and HNEAAG, respectively) in a 2 × 2 factorial design. Cells were preloaded with treatment media containing [15N]-labeled AA for 24 h. The [15N]-labeled media were replaced with treatment media containing [13C]-labeled AA. Media and cells were harvested from plates at 0, 0.5, 1, 5, 15, 30, 60, and 240 min after application of the [13C]-labeled AA and assessed for [15N]- and [13C]-AA label concentrations. The data were used to derive transport, transamination, irreversible loss, and protein-synthesis fluxes. All Val fluxes, except synthesis of rapidly exchanging tissue protein, increased with the HVal treatment. Interestingly, the rapidly exchanging tissue protein, transamination, and irreversible-loss rate constants decreased with HVal, indicating that the significant flux increases were primarily driven by mass action with the cells resisting the flux increases by downregulating activity. However, the decreases could also reflect saturation of processes that would drive down the mass-action rate constants. This is supported by decreases in the same rate constants for Ile and Leu with HVal. This could be due to either competition for shared transamination and oxidation reactions or a reduction in enzymatic activity. Also, NEAAG did not affect Val fluxes, but influx and efflux rate constants increased for both Val and Leu with HNEAAG, indicating an activating substrate effect. Overall, AA transport rates generally responded concordantly with extracellular concentrations, indicating the transporters are not substrate-saturated within the in vivo range. However, BCAA transamination and oxidation enzymes may be approaching saturation within in vivo ranges. In addition, System L transport activity appeared to be stimulated by as much as 75% with high intracellular concentrations of Ala, Gln, and Gly. High concentrations of Val antagonized transport activity of Ile and Leu by 68% and 15%, respectively, indicating competitive inhibition, but this was only observable at HNEAAG concentrations. The exchange transporters of System L transport 8 of the essential AA that make up approximately 40% of milk protein, so better understanding this transporter is an important step for increased efficiency.
Collapse
Affiliation(s)
- A Hruby Weston
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060.
| | - I A M A Teixeira
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060; Department of Animal, Veterinary, and Food Sciences, University of Idaho, Twin Falls, ID 83303-1827
| | - P S Yoder
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060; Perdue AgriBusiness LLC, Salisbury, MD 21804
| | - T Pilonero
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060
| | - M D Hanigan
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060
| |
Collapse
|
25
|
Zhou Z, Yao Y, Sun Y, Wang X, Huang S, Hou J, Wang L, Wei F. Serum betaine and dimethylglycine in mid-pregnancy and the risk of gestational diabetes mellitus: a case-control study. Endocrine 2024:10.1007/s12020-024-03732-4. [PMID: 38448678 DOI: 10.1007/s12020-024-03732-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/04/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE To investigate the associations of choline, betaine, dimethylglycine (DMG), L-carnitine, and Trimethylamine-N-oxide (TMAO) with the risk of Gestational diabetes mellitus (GDM) as well as the markers of glucose homeostasis. METHODS We performed a case-control study including 200 diagnosed GDM cases and 200 controls matched by maternal age (±2 years) and gestational age (±2 weeks). Concentrations of serum metabolites were measured by the high-performance liquid chromatography - tandem mass spectrometry (HPLC-MS/MS). RESULTS Compared to the control group, GDM group had significantly lower serum betaine concentration and betaine/choline ratio, and higher DMG concentration. Furthermore, decreased betaine concentration and betaine/choline ratio, increased DMG concentration showed significant association with the risk of GDM. In addition, serum betaine concentrations were negatively associated with blood glucose levels at 1-h post-glucose load (OGTT-1h), and both betaine and L-carnitine concentrations were positively associated with 1,5-anhydroglucitol levels. Betaine/choline ratio was negatively associated with OGTT-1h and blood glucose levels at 2-h post-glucose load (OGTT-2h) and serum choline concentrations were negatively associated with fasting blood glucose and positively associated with OGTT-2h. CONCLUSION Decreased serum betaine concentrations and betaine/choline ratio, and elevated DMG concentrations could be significant risk factors for GDM. Furthermore, betaine may be associated with blood glucose regulation and short-term glycemic fluctuations.
Collapse
Affiliation(s)
- Ziqing Zhou
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
| | - Yao Yao
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
| | - Yanan Sun
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Medical Insurance Office of Shenzhen Longgang Central Hospital, Shenzhen, Guangdong Province, China
| | - Xin Wang
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Jiamusi University, Jiamusi, Heilongjiang Province, China
| | - Shang Huang
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Shenzhen Children's Hospital of China Medical University, Shenzhen, Guangdong Province, China
| | - Jianli Hou
- Department of Gynecology and Obstetrics, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
| | - Lijun Wang
- Department of Nutrition, School of Medicine, Jinan University, Guangzhou, Guangdong Province, China.
| | - Fengxiang Wei
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China.
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China.
| |
Collapse
|
26
|
Socha MW, Flis W, Wartęga M. Epigenetic Genome Modifications during Pregnancy: The Impact of Essential Nutritional Supplements on DNA Methylation. Nutrients 2024; 16:678. [PMID: 38474806 PMCID: PMC10934520 DOI: 10.3390/nu16050678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Pregnancy is an extremely stressful period in a pregnant woman's life. Currently, women's awareness of the proper course of pregnancy and its possible complications is constantly growing. Therefore, a significant percentage of women increasingly reach for various dietary supplements during gestation. Some of the most popular substances included in multi-ingredient supplements are folic acid and choline. Those substances are associated with positive effects on fetal intrauterine development and fewer possible pregnancy-associated complications. Recently, more and more attention has been paid to the impacts of specific environmental factors, such as diet, stress, physical activity, etc., on epigenetic modifications, understood as changes occurring in gene expression without the direct alteration of DNA sequences. Substances such as folic acid and choline may participate in epigenetic modifications by acting via a one-carbon cycle, leading to the methyl-group donor formation. Those nutrients may indirectly impact genome phenotype by influencing the process of DNA methylation. This review article presents the current state of knowledge on the use of folic acid and choline supplementation during pregnancy, taking into account their impacts on the maternal-fetal unit and possible pregnancy outcomes, and determining possible mechanisms of action, with particular emphasis on their possible impacts on epigenetic modifications.
Collapse
Affiliation(s)
- Maciej W. Socha
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland;
- Department of Obstetrics and Gynecology, St. Adalbert’s Hospital in Gdańsk, Copernicus Healthcare Entity, Jana Pawła II 50, 80-462 Gdańsk, Poland
| | - Wojciech Flis
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland;
- Department of Obstetrics and Gynecology, St. Adalbert’s Hospital in Gdańsk, Copernicus Healthcare Entity, Jana Pawła II 50, 80-462 Gdańsk, Poland
| | - Mateusz Wartęga
- Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland;
| |
Collapse
|
27
|
Liu CC, Chen L, Cai YW, Chen YF, Liu YM, Zhou YJ, Shao ZM, Yu KD. Targeting EMSY-mediated methionine metabolism is a potential therapeutic strategy for triple-negative breast cancer. Cell Rep Med 2024; 5:101396. [PMID: 38290515 PMCID: PMC10897545 DOI: 10.1016/j.xcrm.2024.101396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/19/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024]
Abstract
Cancer stem cells (CSCs) are the most intractable subpopulation of triple-negative breast cancer (TNBC) cells, which have been associated with a high risk of relapse and poor prognosis. However, eradication of CSCs continues to be difficult. Here, we integrate the multiomics data of a TNBC cohort (n = 360) to identify vital markers of CSCs. We discover that EMSY, inducing a BRCAness phenotype, is preferentially expressed in breast CSCs, promotes ALDH+ cells enrichment, and is positively correlated with poor relapse-free survival. Mechanistically, EMSY competitively binds to the Jmjc domain, which is critical for KDM5B enzyme activity, to reshape methionine metabolism, and to promote CSC self-renewal and tumorigenesis in an H3K4 methylation-dependent manner. Moreover, EMSY accumulation in TNBC cells sensitizes them to PARP inhibitors against bulk cells and methionine deprivation against CSCs. These findings indicate that clinically relevant eradication of CSCs could be achieved with a strategy that targets CSC-specific vulnerabilities in amino acid metabolism.
Collapse
Affiliation(s)
- Cui-Cui Liu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Lie Chen
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yu-Wen Cai
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yu-Fei Chen
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yi-Ming Liu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yu-Jie Zhou
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Ke-Da Yu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.
| |
Collapse
|
28
|
Duo H, Chhabra R, Muthusamy V, Zunjare RU, Hossain F. Assessing sequence variation, haplotype analysis and molecular characterisation of aspartate kinase2 (ask2) gene regulating methionine biosynthesis in diverse maize inbreds. Mol Genet Genomics 2024; 299:7. [PMID: 38349549 DOI: 10.1007/s00438-024-02096-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 11/02/2023] [Indexed: 02/15/2024]
Abstract
Traditional maize grain is deficient in methionine, an essential amino acid required for proper growth and development in humans and poultry birds. Thus, development of high methionine maize (HMM) assumes great significance in alleviating malnutrition through sustainable and cost-effective approach. Of various genetic loci, aspartate kinase2 (ask2) gene plays a pivotal role in regulating methionine accumulation in maize. Here, we sequenced the entire ask2 gene of 5394 bp with 13 exons in five wild and five mutant maize inbreds to understand variation at nucleotide level. Sequence analysis revealed that an SNP in exon-13 caused thymine to adenine transversion giving rise to a favourable mutant allele associated with leucine to glutamine substitution in mutant ASK2 protein. Gene-based diversity analysis with 11 InDel markers grouped 48 diverse inbreds into three major clusters with an average genetic dissimilarity of 0.570 (range, 0.0-0.9). The average major allele frequency, gene diversity and PIC are 0.693, 0.408 and 0.341, respectively. A total of 45 haplotypes of the ask2 gene were identified among the maize inbreds. Evolutionary relationship analysis performed among 22 orthologues grouped them into five major clusters. The number of exons varied from 7 to 17, with length varying from 12 to 495 bp among orthologues. ASK2 protein with 565 amino acids was predicted to be in homo-dimeric state with lysine and tartaric acid as binding ligands. Amino acid kinase and ACT domains were found to be conserved in maize and orthologues. The study depicted the presence of enough genetic diversity in ask2 gene in maize, and development of HMM can be accelerated through introgression of favourable allele of ask2 into the parental lines of elite hybrids using molecular breeding.
Collapse
Affiliation(s)
- Hriipulou Duo
- ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Rashmi Chhabra
- ICAR-Indian Agricultural Research Institute, New Delhi, India
| | | | | | - Firoz Hossain
- ICAR-Indian Agricultural Research Institute, New Delhi, India.
| |
Collapse
|
29
|
Chen G, Zhou G, Zhai L, Bao X, Tiwari N, Li J, Mottillo E, Wang J. SHMT2 reduces fatty liver but is necessary for liver inflammation and fibrosis in mice. Commun Biol 2024; 7:173. [PMID: 38347107 PMCID: PMC10861579 DOI: 10.1038/s42003-024-05861-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/26/2024] [Indexed: 02/15/2024] Open
Abstract
Non-alcoholic fatty liver disease is associated with an irregular serine metabolism. Serine hydroxymethyltransferase 2 (SHMT2) is a liver enzyme that breaks down serine into glycine and one-carbon (1C) units critical for liver methylation reactions and overall health. However, the contribution of SHMT2 to hepatic 1C homeostasis and biological functions has yet to be defined in genetically modified animal models. We created a mouse strain with targeted SHMT2 knockout in hepatocytes to investigate this. The absence of SHMT2 increased serine and glycine levels in circulation, decreased liver methylation potential, and increased susceptibility to fatty liver disease. Interestingly, SHMT2-deficient mice developed simultaneous fatty liver, but when fed a diet high in fat, fructose, and cholesterol, they had significantly less inflammation and fibrosis. This study highlights the critical role of SHMT2 in maintaining hepatic 1C homeostasis and its stage-specific functions in the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Guohua Chen
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, 48202, USA
| | - Guoli Zhou
- Biomedical Research Informatics Core, Clinical and Translational Sciences Institute, Michigan State University, East Lansing, MI, 48824, USA
| | - Lidong Zhai
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xun Bao
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, 48202, USA
| | - Nivedita Tiwari
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Jing Li
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, 48202, USA
| | - Emilio Mottillo
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI, 48202, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48202, USA
| | - Jian Wang
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, 48202, USA.
| |
Collapse
|
30
|
Liu J, Han L, Hou S, Gui L, Yuan Z, Sun S, Wang Z, Yang B. Integrated metabolome and microbiome analysis reveals the effect of rumen-protected sulfur-containing amino acids on the meat quality of Tibetan sheep meat. Front Microbiol 2024; 15:1345388. [PMID: 38389537 PMCID: PMC10883651 DOI: 10.3389/fmicb.2024.1345388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/17/2024] [Indexed: 02/24/2024] Open
Abstract
Introduction This study investigated the effects of rumen-protected sulfur-containing amino acids (RPSAA) on the rumen and jejunal microbiota as well as on the metabolites and meat quality of the longissimus lumborum (LL) in Tibetan sheep. Methods By combining 16S rDNA sequencing with UHPLC-Q-TOF MS and Pearson correlation analysis, the relationship between gastrointestinal microbiota, muscle metabolites and meat quality was identified. Results The results showed that feeding RPSAA can increase the carcass weight, abdominal fat thickness (AP-2 group), and back fat thickness (AP-2 and AP-3 group) of Tibetan sheep. The water holding capacity (WHC), texture, and shear force (SF) of LL in the two groups also increased although the fatty acids content and brightness (L*) value significantly decreased in the AP-2 group. Metabolomics and correlation analysis further showed that RPSAA could significantly influence the metabolites in purine metabolism, thereby affecting L* and SF. In addition, RPSAA was beneficial for the fermentation of the rumen and jejunum. In both groups, the abundance of Prevotella 1, Lachnospiraceae NK3A20 group, Prevotella UCG-003, Lachnospiraceae ND3007 group in the rumen as well as the abundance of Eubacterium nodatum group and Mogibacterium group in the jejunum increased. In contrast, that of Turicibacter pathogens in the jejunum was reduced. The above microorganisms could regulate meat quality by regulating the metabolites (inosine, hypoxanthine, linoleic acid, palmitic acid, etc.) in purine and fatty acids metabolism. Discussion Overall, reducing the levels of crude proteins in the diet and feeding RPSAA is likely to improve the carcass quality of Tibetan sheep, with the addition of RPMET (AP-2) yielding the best edible quality, possibly due to its ability to influence the gastrointestinal microbiota to subsequently regulate muscle metabolites.
Collapse
Affiliation(s)
- JiQian Liu
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| | - Lijuan Han
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| | - Shengzhen Hou
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| | - Linsheng Gui
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| | - Zhenzhen Yuan
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| | - Shengnan Sun
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| | - Zhiyou Wang
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| | - Baochun Yang
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| |
Collapse
|
31
|
Zhou L. Homocysteine and Parkinson's disease. CNS Neurosci Ther 2024; 30:e14420. [PMID: 37641911 PMCID: PMC10848096 DOI: 10.1111/cns.14420] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023] Open
Abstract
Homocysteine (Hcy) is an important metabolite in methionine metabolism. When the metabolic pathway of homocysteine is abnormal, it will accumulate in the body and eventually lead to hyperhomocysteinemia. In recent years, many studies have found that hyperhomocysteinemia is related to the occurrence and development of Parkinson's disease. This study reviews the roles of homocysteine in the pathogenesis of Parkinson's disease and illustrates the harmful effects of hyperhomocysteinemia on Parkinson's disease.
Collapse
Affiliation(s)
- Lingyan Zhou
- Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| |
Collapse
|
32
|
Takai S, Lee H, Kim M, Torii S, Nishihara K, Oh J, Masaki T, Ikuta K, Iwamoto E, Masuda K, Uemoto Y, Terada F, Haga S, Roh S. Dynamics of blood Taurine concentration and its correlation with nutritional and physiological status during the fattening period of Japanese black cattle. J Anim Sci 2024; 102:skae347. [PMID: 39535934 PMCID: PMC11631192 DOI: 10.1093/jas/skae347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024] Open
Abstract
Taurine, biosynthesized from methionine or cysteine in the liver, plays a crucial regulatory role in bile acid conjugation, antioxidant effects, and glucose and cholesterol metabolism. This may influence the metabolic changes associated with fat accumulation in beef cattle. However, the physiological role of taurine in this species has not been fully elucidated. In this study, we explored the physiological role of taurine in Japanese Black steers (Bos taurus) in different phases during the fattening period. To examine the correlation among plasma taurine concentrations, various physiological parameters, and genes related to taurine synthesis in the liver, we used biopsied liver tissues, blood samples, and rumen fluids collected from 21 steers at three different stages, i.e., early (T1; 13 mo of age), middle (T2; 20 mo of age), and late (T3; 28 mo of age) phases. Additionally, to investigate the regulatory mechanisms underlying the expression profile of taurine synthesis genes, primary bovine hepatocytes obtained from 4-wk-old Holstein calves were treated with palmitate, oleate, acetate, propionate, or β-hydroxybutyrate (BHBA). Plasma taurine and cholesterol concentrations significantly (P < 0.001) increased in the T2 phase, which is potentially attributable to increased energy intake and assimilation induced by increased intake of concentrated feed. Cysteine-sulfinic acid decarboxylase (CSAD) expression significantly increased (P < 0.01) in T2 than in other phases. The expression levels of cysteine dioxygenase type 1 (CDO1) and cholesterol 7 alpha-hydroxylase (CYP7A1) were significantly higher (P < 0.05) in T2 than in T3; moreover, the CDO1/glutamate-cysteine ligase catalytic subunit (GCLC) ratio was higher (P < 0.05) in T2 than in T1. Plasma taurine concentrations were positively correlated with plasma methionine (r = 0.51; P < 0.05) and total cholesterol (r = 0.56; P < 0.05) concentrations at T2. Relative CDO1 mRNA expression was upregulated in cultured bovine hepatocytes treated with oleate and propionate, whereas it was downregulated upon acetate treatment. These findings indicate that the increase in plasma taurine concentrations in the T2 phase is associated with changes in lipid and methionine metabolism in Japanese Black steers.
Collapse
Affiliation(s)
- Shuntaro Takai
- Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Huseong Lee
- Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Minji Kim
- Department of Animal Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Shinichiro Torii
- Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Koki Nishihara
- Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Joonpyo Oh
- Strategic Marketing and Technology, Cargill Animal Nutrition, Seongnam 13630, Republic of Korea
| | - Tatsunori Masaki
- Hyogo Prefectural Technology Center of Agriculture, Forestry and Fisheries, Kasai, Hyogo, 679-0198, Japan
| | - Kentaro Ikuta
- Hyogo Prefectural Technology Center of Agriculture, Forestry and Fisheries, Kasai, Hyogo, 679-0198, Japan
| | - Eiji Iwamoto
- Hyogo Prefectural Technology Center of Agriculture, Forestry and Fisheries, Kasai, Hyogo, 679-0198, Japan
| | - Kota Masuda
- Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Yoshinobu Uemoto
- Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Fuminori Terada
- Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Satoshi Haga
- Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Sanggun Roh
- Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| |
Collapse
|
33
|
Vlasova VV, Shmagel KV. T Lymphocyte Metabolic Features and Techniques to Modulate Them. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1857-1873. [PMID: 38105204 DOI: 10.1134/s0006297923110159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/21/2023] [Accepted: 08/27/2023] [Indexed: 12/19/2023]
Abstract
T cells demonstrate high degree of complexity and broad range of functions, which distinguish them from other immune cells. Throughout their lifetime, T lymphocytes experience several functional states: quiescence, activation, proliferation, differentiation, performance of effector and regulatory functions, memory formation, and apoptosis. Metabolism supports all functions of T cells, providing lymphocytes with energy, biosynthetic substrates, and signaling molecules. Therefore, T cells usually restructure their metabolism as they transition from one functional state to another. Strong association between the metabolism and T cell functions implies that the immune response can be controlled by manipulating metabolic processes within T lymphocytes. This review aims to highlight the main metabolic adaptations necessary for the T cell function, as well as the recent progress in techniques to modulate metabolic features of lymphocytes.
Collapse
Affiliation(s)
- Violetta V Vlasova
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, 614081, Perm, Russia.
| | - Konstantin V Shmagel
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, 614081, Perm, Russia
| |
Collapse
|
34
|
Espe M, Adam AC, Saito T, Skjærven KH. Methionine: An Indispensable Amino Acid in Cellular Metabolism and Health of Atlantic Salmon. AQUACULTURE NUTRITION 2023; 2023:5706177. [PMID: 37927379 PMCID: PMC10624553 DOI: 10.1155/2023/5706177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023]
Abstract
Methionine is an indispensable amino acid with an important role as the main methyl donor in cellular metabolism for both fish and mammals. Metabolization of methionine to the methyl donor S-adenosylmethionine (SAM) has consequence for polyamine, carnitine, phospholipid, and creatine synthesis as well as epigenetic modifications such as DNA- and histone tail methylation. Methionine can also be converted to cysteine and contributes as a precursor for taurine and glutathione synthesis. Moreover, methionine is the start codon for every protein being synthetized and thereby serves an important role in initiating translation. Modern salmon feed is dominated by plant ingredients containing less taurine, carnitine, and creatine than animal-based ingredients. This shift results in competition for SAM due to an increasing need to endogenously synthesize associated metabolites. The availability of methionine has profound implications for various metabolic pathways including allosteric regulation. This necessitates a higher nutritional need to meet the requirement as a methyl donor, surpassing the quantities for protein synthesis and growth. This comprehensive review provides an overview of the key metabolic pathways in which methionine plays a central role as methyl donor and unfolds the implications for methylation capacity, metabolism, and overall health particularly emphasizing the development of fatty liver, oxidation, and inflammation when methionine abundance is insufficient focusing on nutrition for Atlantic salmon (Salmo salar).
Collapse
Affiliation(s)
- M. Espe
- Institute of Marine Research, P.O. Box 5817 Nordnes, Bergen, Norway
| | - A. C. Adam
- Institute of Marine Research, P.O. Box 5817 Nordnes, Bergen, Norway
| | - T. Saito
- Institute of Marine Research, P.O. Box 5817 Nordnes, Bergen, Norway
| | - K. H. Skjærven
- Institute of Marine Research, P.O. Box 5817 Nordnes, Bergen, Norway
| |
Collapse
|
35
|
Su D, Zhang R, Wang X, Ding Q, Che F, Liu Z, Xu J, Zhao Y, Ji K, Wu W, Yan C, Li P, Tang B. Shedding Light on Lysosomal Malondialdehyde Affecting Vitamin B 12 Transport during Cerebral Ischemia/Reperfusion Injury. J Am Chem Soc 2023; 145:22609-22619. [PMID: 37803879 DOI: 10.1021/jacs.3c07809] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) is often accompanied by upregulation of homocysteine (Hcy). Excessive Hcy damages cerebral vascular endothelial cells and neurons, inducing neurotoxicity and even neurodegeneration. Normally, supplementation of vitamin B12 is an ideal intervention to reduce Hcy. However, vitamin B12 therapy is clinically inefficacious for CIRI. Considering oxidative stress is closely related to CIRI, the lysosome is the pivotal site for vitamin B12 transport. Lysosomal oxidative stress might hinder the transport of vitamin B12. Whether lysosomal malondialdehyde (lysosomal MDA), as the authoritative biomarker of lysosomal oxidative stress, interferes with the transport of vitamin B12 has not been elucidated. This is ascribed to the absence of effective methods for real-time and in situ measurement of lysosomal MDA within living brains. Herein, a fluorescence imaging agent, Lyso-MCBH, was constructed to specifically monitor lysosomal MDA by entering the brain and targeting the lysosome. Erupting the lysosomal MDA level in living brains of mice under CIRI was first observed using Lyso-MCBH. Excessive lysosomal MDA was found to affect the efficacy of vitamin B12 by blocking the transport of vitamin B12 from the lysosome to the cytoplasm. More importantly, the expression and function of the vitamin B12 transporter LMBD1 were proved to be associated with excessive lysosomal MDA. Altogether, the revealing of the lysosomal MDA-LMBD1 axis provides a cogent interpretation of the inefficacy of vitamin B12 in CIRI, which could be a prospective therapeutic target.
Collapse
Affiliation(s)
- Di Su
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, Shandong, People's Republic of China
| | - Ran Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, Shandong, People's Republic of China
| | - Xin Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, Shandong, People's Republic of China
| | - Qi Ding
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, Shandong, People's Republic of China
| | - Feida Che
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, Shandong, People's Republic of China
| | - Zhenzhen Liu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, Shandong, People's Republic of China
| | - Jingwen Xu
- Department of Neurology, Qi-Lu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qi-Lu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Yuying Zhao
- Department of Neurology, Qi-Lu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qi-Lu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Kunqian Ji
- Department of Neurology, Qi-Lu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qi-Lu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Wei Wu
- Department of Neurology, Qi-Lu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Brain Science Research Institute, Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Chuanzhu Yan
- Department of Neurology, Qi-Lu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qi-Lu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Brain Science Research Institute, Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, Shandong, People's Republic of China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, Shandong, People's Republic of China
- Laoshan Laboratory, Qingdao 266237, Shandong, People's Republic of China
| |
Collapse
|
36
|
Perez MF, Sarkies P. Histone methyltransferase activity affects metabolism in human cells independently of transcriptional regulation. PLoS Biol 2023; 21:e3002354. [PMID: 37883365 PMCID: PMC10602318 DOI: 10.1371/journal.pbio.3002354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
The N-terminal tails of eukaryotic histones are frequently posttranslationally modified. The role of these modifications in transcriptional regulation is well-documented. However, the extent to which the enzymatic processes of histone posttranslational modification might affect metabolic regulation is less clear. Here, we investigated how histone methylation might affect metabolism using metabolomics, proteomics, and RNA-seq data from cancer cell lines, primary tumour samples and healthy tissue samples. In cancer, the expression of histone methyltransferases (HMTs) was inversely correlated to the activity of NNMT, an enzyme previously characterised as a methyl sink that disposes of excess methyl groups carried by the universal methyl donor S-adenosyl methionine (SAM or AdoMet). In healthy tissues, histone methylation was inversely correlated to the levels of an alternative methyl sink, PEMT. These associations affected the levels of multiple histone marks on chromatin genome-wide but had no detectable impact on transcriptional regulation. We show that HMTs with a variety of different associations to transcription are co-regulated by the Retinoblastoma (Rb) tumour suppressor in human cells. Rb-mutant cancers show increased total HMT activity and down-regulation of NNMT. Together, our results suggest that the total activity of HMTs affects SAM metabolism, independent of transcriptional regulation.
Collapse
Affiliation(s)
- Marcos Francisco Perez
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Department of Cells and Tissues, Instituto de Biologia Molecular de Barcelona (IBMB), CSIC, Barcelona, Spain
| | - Peter Sarkies
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
37
|
Jiachen Z, Paul Kwong Hang T, Kenneth Kak Yuen W, Vincent Chi Hang L. Pathological role of methionine in the initiation and progression of biliary atresia. Front Pediatr 2023; 11:1263836. [PMID: 37772039 PMCID: PMC10522914 DOI: 10.3389/fped.2023.1263836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/21/2023] [Indexed: 09/30/2023] Open
Abstract
Methionine (Met) is an essential amino acid, and its excessive dietary intake and/or its metabolism disturbance could lead to accumulation/depletion of hepatic Met and some of the key intermediates of these pathways, which would interfere normal liver function and would be associated with liver diseases. Biliary atresia (BA) is a life-threatening disease characterized by inflammatory fibrosclerosing changes of the intrahepatic and extrahepatic biliary systems and is the primary cause of obstructive neonatal cholestasis with a rapid course of liver failure. However, its pathogenesis remains unknown. Previous studies reported elevated Met level in patients with obstructive cholestasis, suggesting a potential link between Met and BA. This paper reviews the Met metabolism in normal conditions and its dysregulation under abnormal conditions, the possible causes of hypermethioninemia, and its connection to BA pathogenesis: Abnormal hepatic level of Met could lead to a perturbation of redox homeostasis and mitochondrial functions of hepatocytes, enhancement of viral infectivity, and dysregulation of innate and adaptative immune cells in response to infection/damage of the liver contributing to the initiation/progression of BA.
Collapse
Affiliation(s)
- Zheng Jiachen
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Tam Paul Kwong Hang
- Faculty of Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Wong Kenneth Kak Yuen
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Surgery, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Lui Vincent Chi Hang
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
38
|
Tsai PY, Shui B, Lee S, Liu Y, Qu Y, Cheng C, Edwards K, Wong C, Meng-Killeen R, Soloway PD, Barrow JJ. Ado-Mediated Depletion of Taurine Impairs Mitochondrial Respiratory Capacity and Alters the Chromatin Landscape of Inguinal Adipose Tissue. Nutrients 2023; 15:3532. [PMID: 37630723 PMCID: PMC10458711 DOI: 10.3390/nu15163532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Non-shivering thermogenesis (NST) has strong potential to combat obesity; however, a safe molecular approach to activate this process has not yet been identified. The sulfur amino acid taurine has the ability to safely activate NST and confer protection against obesity and metabolic disease in both mice and humans, but the mechanism of this action is unknown. In this study, we discover that a suite of taurine biosynthetic enzymes, especially that of cysteamine dioxygenase (ADO), significantly increases in response to β3 adrenergic signaling in inguinal adipose tissue (IWAT) in order to increase intracellular concentrations of taurine. We further show that ADO is critical for thermogenic mitochondrial respiratory function as its ablation in adipocytes significantly reduces taurine levels, which leads to declines in mitochondrial oxygen consumption rates. Finally, we demonstrate via assay for transposase-accessible chromatin with sequencing (ATAC-seq) that taurine supplementation in beige adipocytes has the ability to remodel the chromatin landscape to increase the chromatin accessibility and transcription of genes, such as glucose-6-phosphate isomerase 1 (Gpi1), which are critical for NST. Taken together, our studies highlight a potential mechanism for taurine in the activation of NST that can be leveraged toward the treatment of obesity and metabolic disease.
Collapse
Affiliation(s)
- Pei-Yin Tsai
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA; (P.-Y.T.)
| | - Bo Shui
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14850, USA
| | - Seoyeon Lee
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA; (P.-Y.T.)
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14850, USA
| | - Yang Liu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA; (P.-Y.T.)
| | - Yue Qu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA; (P.-Y.T.)
| | - Chloe Cheng
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14850, USA
| | - Kaydine Edwards
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA; (P.-Y.T.)
| | - Callie Wong
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA; (P.-Y.T.)
| | - Ryan Meng-Killeen
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA; (P.-Y.T.)
| | - Paul D. Soloway
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA; (P.-Y.T.)
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14850, USA
| | - Joeva J. Barrow
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA; (P.-Y.T.)
| |
Collapse
|
39
|
Perez M, Aroh O, Sun Y, Lan Y, Juniper SK, Young CR, Angers B, Qian PY. Third-Generation Sequencing Reveals the Adaptive Role of the Epigenome in Three Deep-Sea Polychaetes. Mol Biol Evol 2023; 40:msad172. [PMID: 37494294 PMCID: PMC10414810 DOI: 10.1093/molbev/msad172] [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: 02/24/2023] [Revised: 06/16/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023] Open
Abstract
The roles of DNA methylation in invertebrates are poorly characterized, and critical data are missing for the phylum Annelida. We fill this knowledge gap by conducting the first genome-wide survey of DNA methylation in the deep-sea polychaetes dominant in deep-sea vents and seeps: Paraescarpia echinospica, Ridgeia piscesae, and Paralvinella palmiformis. DNA methylation calls were inferred from Oxford Nanopore sequencing after assembling high-quality genomes of these animals. The genomes of these worms encode all the key enzymes of the DNA methylation metabolism and possess a mosaic methylome similar to that of other invertebrates. Transcriptomic data of these polychaetes support the hypotheses that gene body methylation strengthens the expression of housekeeping genes and that promoter methylation acts as a silencing mechanism but not the hypothesis that DNA methylation suppresses the activity of transposable elements. The conserved epigenetic profiles of genes responsible for maintaining homeostasis under extreme hydrostatic pressure suggest DNA methylation plays an important adaptive role in these worms.
Collapse
Affiliation(s)
- Maeva Perez
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, China
- Department of Biological Sciences, Université de Montréal, Montréal, Canada
| | - Oluchi Aroh
- Department of Biological Sciences, Auburn University, Auburn, AL, USA
| | - Yanan Sun
- Laboratory of Marine Organism Taxonomy and Phylogeny, Chinese Academy of Sciences, Institute of Oceanology, Qingdao, China
| | - Yi Lan
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, China
| | - Stanley Kim Juniper
- School of Earth and Ocean Sciences, University of Victoria, Victoria, Canada
| | | | - Bernard Angers
- Department of Biological Sciences, Université de Montréal, Montréal, Canada
| | - Pei-Yuan Qian
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, China
| |
Collapse
|
40
|
Wan L, Zhu S, Chen Z, Qiu R, Tang B, Jiang H. Multidimensional biomarkers for multiple system atrophy: an update and future directions. Transl Neurodegener 2023; 12:38. [PMID: 37501056 PMCID: PMC10375766 DOI: 10.1186/s40035-023-00370-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023] Open
Abstract
Multiple system atrophy (MSA) is a fatal progressive neurodegenerative disease. Biomarkers are urgently required for MSA to improve the diagnostic and prognostic accuracy in clinic and facilitate the development and monitoring of disease-modifying therapies. In recent years, significant research efforts have been made in exploring multidimensional biomarkers for MSA. However, currently few biomarkers are available in clinic. In this review, we systematically summarize the latest advances in multidimensional biomarkers for MSA, including biomarkers in fluids, tissues and gut microbiota as well as imaging biomarkers. Future directions for exploration of novel biomarkers and promotion of implementation in clinic are also discussed.
Collapse
Affiliation(s)
- Linlin Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, 410008, China
| | - Sudan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
| | - Rong Qiu
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China.
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, 410008, China.
| |
Collapse
|
41
|
Li W, Jia Y, Gong Z, Dong Z, Yu F, Fu Y, Jiang C, Kong W. Ablation of the gut microbiota alleviates high-methionine diet-induced hyperhomocysteinemia and glucose intolerance in mice. NPJ Sci Food 2023; 7:36. [PMID: 37460578 DOI: 10.1038/s41538-023-00212-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 07/12/2023] [Indexed: 07/20/2023] Open
Abstract
A high-methionine (HM) diet leads to hyperhomocysteinemia (HHcy), while gastrointestinal tissue is an important site of net homocysteine (Hcy) production. However, the role of the gut microbiota in host HHcy remains obscure. This study aimed to determine whether gut microbiota ablation could alleviate host HHcy and glucose intolerance and reveal the underlying mechanism. The results showed that the HM diet-induced HHcy and glucose intolerance in mice, while antibiotic administration decreased the plasma level of Hcy and reversed glucose intolerance. HM diet increased intestinal epithelial homocysteine levels, while antibiotic treatment decreased intestinal epithelial homocysteine levels under the HM diet. Gut microbiota depletion had no effect on the gene expression and enzyme activity of CBS and BHMT in the livers of HM diet-fed mice. The HM diet altered the composition of the gut microbiota with marked increases in the abundances of Faecalibaculum and Dubosiella, which were also positively correlated with plasma Hcy concentrations. An in-depth analysis of the bacterial cysteine and methionine metabolism pathways showed that the abundances of two homocysteine biosynthesis-related KEGG orthologies (KOs) were markedly increased in the gut microbiota in HM diet-fed mice. Hcy was detected from Dubosiella newyorkensis-cultured supernatant by liquid chromatography-tandem mass spectrometry (LC‒MS) analysis. In conclusion, these findings suggested that the HM diet-induced HHcy and glucose intolerance in mice, by reshaping the composition of the gut microbiota, which might produce and secrete Hcy.
Collapse
Affiliation(s)
- Wenqiang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yiting Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhao Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
42
|
Liu DD, Zhang CY, Zhang JT, Gu LM, Xu GT, Zhang JF. Epigenetic modifications and metabolic memory in diabetic retinopathy: beyond the surface. Neural Regen Res 2023; 18:1441-1449. [PMID: 36571340 PMCID: PMC10075108 DOI: 10.4103/1673-5374.361536] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/10/2022] [Accepted: 10/26/2022] [Indexed: 11/19/2022] Open
Abstract
Epigenetics focuses on DNA methylation, histone modification, chromatin remodeling, noncoding RNAs, and other gene regulation mechanisms beyond the DNA sequence. In the past decade, epigenetic modifications have drawn more attention as they participate in the development and progression of diabetic retinopathy despite tight control of glucose levels. The underlying mechanisms of epigenetic modifications in diabetic retinopathy still urgently need to be elucidated. The diabetic condition facilitates epigenetic changes and influences target gene expression. In this review, we summarize the involvement of epigenetic modifications and metabolic memory in the development and progression of diabetic retinopathy and propose novel insights into the treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- Dan-Dan Liu
- Department of Ophthalmology of Tongji Hospital, Tongji Eye Institute, Department of Regenerative Medicine, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Chao-Yang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Jing-Ting Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Li-Min Gu
- Department of Ophthalmology, Shanghai Aier Eye Hospital, Shanghai, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Tongji Hospital, Tongji Eye Institute, Department of Regenerative Medicine, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Jing-Fa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
43
|
Li Z, Liu Q, Sun J, Sun J, Li M, Zhang Y, Deng A, Liu S, Wen T. Multivariate modular metabolic engineering for enhanced L-methionine biosynthesis in Escherichia coli. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2023; 16:101. [PMID: 37312226 DOI: 10.1186/s13068-023-02347-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/23/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND L-Methionine is the only bulk amino acid that has not been industrially produced by the fermentation method. Due to highly complex and strictly regulated biosynthesis, the development of microbial strains for high-level L-methionine production has remained challenging in recent years. RESULTS By strengthening the L-methionine terminal synthetic module via site-directed mutation of L-homoserine O-succinyltransferase (MetA) and overexpression of metAfbr, metC, and yjeH, L-methionine production was increased to 1.93 g/L in shake flask fermentation. Deletion of the pykA and pykF genes further improved L-methionine production to 2.51 g/L in shake flask fermentation. Computer simulation and auxotrophic experiments verified that during the synthesis of L-methionine, equimolar amounts of L-isoleucine were accumulated via the elimination reaction of cystathionine γ-synthetase MetB due to the insufficient supply of L-cysteine. To increase the supply of L-cysteine, the L-cysteine synthetic module was strengthened by overexpression of cysEfbr, serAfbr, and cysDN, which further increased the production of L-methionine by 52.9% and significantly reduced the accumulation of the byproduct L-isoleucine by 29.1%. After optimizing the addition of ammonium thiosulfate, the final metabolically engineered strain MET17 produced 21.28 g/L L-methionine in 64 h with glucose as the carbon source in a 5 L fermenter, representing the highest L-methionine titer reported to date. CONCLUSIONS In this study, a high-efficiency strain for L-methionine production was derived from wild-type Escherichia coli W3110 by rational metabolic engineering strategies, providing an efficient platform for the industrial production of L-methionine.
Collapse
Affiliation(s)
- Zhongcai Li
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qian Liu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiahui Sun
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Jianjian Sun
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mingjie Li
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yun Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Aihua Deng
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuwen Liu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Tingyi Wen
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
- China Innovation Academy for Green Manufacture, Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
44
|
Ivanov AV, Popov MA, Metelkin AA, Aleksandrin VV, Agafonov EG, Kruglova MP, Silina EV, Stupin VA, Maslennikov RA, Kubatiev AA. Influence of Coronary Artery Bypass Grafts on Blood Aminothiols in Patients with Coronary Artery Disease. Metabolites 2023; 13:743. [PMID: 37367901 PMCID: PMC10305081 DOI: 10.3390/metabo13060743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/03/2023] [Accepted: 06/08/2023] [Indexed: 06/28/2023] Open
Abstract
Coronary artery disease (CAD) and the coronary artery bypass graft (CABG) are associated with a decreased blood glutathione (bGSH) level. Since GSH metabolism is closely related to other aminothiols (homocysteine and cysteine) and glucose, the aim of this study was to reveal the associations of bGSH with glucose and plasma aminothiols in CAD patients (N = 35) before CABG and in the early postoperative period. Forty-three volunteers with no history of cardiovascular disease formed the control group. bGSH and its redox status were significantly lower in CAD patients at admission. CABG had no significant effect on these parameters, with the exception of an increase in the bGSH/hemoglobin ratio. At admission, CAD patients were characterized by negative associations of homocysteine and cysteine with bGSH. All these associations disappeared after CABG. An association was found between an increase in oxidized GSH in the blood in the postoperative period and fasting glucose levels. Thus, CAD is associated with the depletion of the intracellular pool and the redox status of bGSH, in which hyperhomocysteinemia and a decrease in the bioavailability of the extracellular pool of cysteine play a role. The present study indicates that CABG causes disruptions in aminothiol metabolism and induces the synthesis of bGSH. Moreover, glucose becomes an important factor in the dysregulation of GSH metabolism in CABG.
Collapse
Affiliation(s)
- Alexander Vladimirovich Ivanov
- Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia; (A.A.M.); (V.V.A.); (M.P.K.); (A.A.K.)
| | - Mikhail Aleksandrovich Popov
- Moscow Regional Research and Clinical Institute n.a. M.F. Vladimirskiy, Shchepkin St., 61/2, 129110 Moscow, Russia; (M.A.P.); (E.G.A.); (R.A.M.)
| | - Arkady Andreevich Metelkin
- Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia; (A.A.M.); (V.V.A.); (M.P.K.); (A.A.K.)
| | - Valery Vasil’evich Aleksandrin
- Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia; (A.A.M.); (V.V.A.); (M.P.K.); (A.A.K.)
| | - Evgeniy Gennad’evich Agafonov
- Moscow Regional Research and Clinical Institute n.a. M.F. Vladimirskiy, Shchepkin St., 61/2, 129110 Moscow, Russia; (M.A.P.); (E.G.A.); (R.A.M.)
| | - Maria Petrovna Kruglova
- Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia; (A.A.M.); (V.V.A.); (M.P.K.); (A.A.K.)
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya St., 8, 119991 Moscow, Russia;
| | - Ekaterina Vladimirovna Silina
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya St., 8, 119991 Moscow, Russia;
| | - Victor Aleksandrovich Stupin
- Department of Hospital Surgery No. 1, Pirogov Russian National Research Medical University, Ostrovityanova St., 1, 117997 Moscow, Russia;
| | - Ruslan Andreevich Maslennikov
- Moscow Regional Research and Clinical Institute n.a. M.F. Vladimirskiy, Shchepkin St., 61/2, 129110 Moscow, Russia; (M.A.P.); (E.G.A.); (R.A.M.)
| | - Aslan Amirkhanovich Kubatiev
- Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia; (A.A.M.); (V.V.A.); (M.P.K.); (A.A.K.)
| |
Collapse
|
45
|
Katarachia SA, Markaki SP, Velentzas AD, Stravopodis DJ. Genetic Targeting of dSAMTOR, A Negative dTORC1 Regulator, during Drosophila Aging: A Tissue-Specific Pathology. Int J Mol Sci 2023; 24:ijms24119676. [PMID: 37298625 DOI: 10.3390/ijms24119676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/23/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
mTORC1 regulates mammalian cell metabolism and growth in response to diverse environmental stimuli. Nutrient signals control the localization of mTORC1 onto lysosome surface scaffolds that are critically implicated in its amino acid-dependent activation. Arginine, leucine and S-adenosyl-methionine (SAM) can serve as major mTORC1-signaling activators, with SAM binding to SAMTOR (SAM + TOR), a fundamental SAM sensor, preventing the protein's (SAMTOR's) inhibitory action(s) against mTORC1, thereby triggering its (mTORC1) kinase activity. Given the lack of knowledge regarding the role of SAMTOR in invertebrates, we have identified the Drosophila SAMTOR homologue (dSAMTOR) in silico and have, herein, genetically targeted it through the utilization of the GAL4/UAS transgenic tool. Survival profiles and negative geotaxis patterns were examined in both control and dSAMTOR-downregulated adult flies during aging. One of the two gene-targeted schemes resulted in lethal phenotypes, whereas the other one caused rather moderate pathologies in most tissues. The screening of head-specific kinase activities, via PamGene technology application, unveiled the significant upregulation of several kinases, including the dTORC1 characteristic substrate dp70S6K, in dSAMTOR-downregulated flies, thus strongly supporting the inhibitory dSAMTOR action(s) upon the dTORC1/dp70S6K signaling axis in Drosophila brain settings. Importantly, genetic targeting of the Drosophila BHMT bioinformatics counterpart (dBHMT), an enzyme that catabolizes betaine to produce methionine (the SAM precursor), led to severe compromises in terms of fly longevity, with glia-, motor neuron- and muscle-specific dBHMT downregulations exhibiting the strongest effects. Abnormalities in wing vein architectures were also detected in dBHMT-targeted flies, thereby justifying their notably reduced negative geotaxis capacities herein observed mainly in the brain-(mid)gut axis. In vivo adult fly exposure to clinically relevant doses of methionine revealed the mechanistic synergism of decreased dSAMTOR and increased methionine levels in pathogenic longevity, thus rendering (d)SAMTOR an important component in methionine-associated disorders, including homocystinuria(s).
Collapse
Affiliation(s)
- Stamatia A Katarachia
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), 15701 Athens, Greece
| | - Sophia P Markaki
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), 15701 Athens, Greece
| | - Athanassios D Velentzas
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), 15701 Athens, Greece
| | - Dimitrios J Stravopodis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), 15701 Athens, Greece
| |
Collapse
|
46
|
Wünsch AC, Ries E, Heinzelmann S, Frabschka A, Wagner PC, Rauch T, Koderer C, El-Mesery M, Volland JM, Kübler AC, Hartmann S, Seher A. Metabolic Silencing via Methionine-Based Amino Acid Restriction in Head and Neck Cancer. Curr Issues Mol Biol 2023; 45:4557-4573. [PMID: 37367038 DOI: 10.3390/cimb45060289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
In recent years, various forms of caloric restriction (CR) and amino acid or protein restriction (AAR or PR) have shown not only success in preventing age-associated diseases, such as type II diabetes and cardiovascular diseases, but also potential for cancer therapy. These strategies not only reprogram metabolism to low-energy metabolism (LEM), which is disadvantageous for neoplastic cells, but also significantly inhibit proliferation. Head and neck squamous cell carcinoma (HNSCC) is one of the most common tumour types, with over 600,000 new cases diagnosed annually worldwide. With a 5-year survival rate of approximately 55%, the poor prognosis has not improved despite extensive research and new adjuvant therapies. Therefore, for the first time, we analysed the potential of methionine restriction (MetR) in selected HNSCC cell lines. We investigated the influence of MetR on cell proliferation and vitality, the compensation for MetR by homocysteine, the gene regulation of different amino acid transporters, and the influence of cisplatin on cell proliferation in different HNSCC cell lines.
Collapse
Affiliation(s)
- Anna Chiara Wünsch
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Elena Ries
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Sina Heinzelmann
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Andrea Frabschka
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Peter Christoph Wagner
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Theresa Rauch
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Corinna Koderer
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Mohamed El-Mesery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Julian Manuel Volland
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Alexander Christian Kübler
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Stefan Hartmann
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Axel Seher
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| |
Collapse
|
47
|
Fang W, Jiang L, Zhu Y, Yang S, Qiu H, Cheng J, Liang Q, Tu ZC, Ye C. Methionine restriction constrains lipoylation and activates mitochondria for nitrogenic synthesis of amino acids. Nat Commun 2023; 14:2504. [PMID: 37130856 PMCID: PMC10154411 DOI: 10.1038/s41467-023-38289-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 04/21/2023] [Indexed: 05/04/2023] Open
Abstract
Methionine restriction (MR) provides metabolic benefits in many organisms. However, mechanisms underlying the MR-induced effect remain incompletely understood. Here, we show in the budding yeast S. cerevisiae that MR relays a signal of S-adenosylmethionine (SAM) deprivation to adapt bioenergetic mitochondria to nitrogenic anabolism. In particular, decreases in cellular SAM constrain lipoate metabolism and protein lipoylation required for the operation of the tricarboxylic acid (TCA) cycle in the mitochondria, leading to incomplete glucose oxidation with an exit of acetyl-CoA and α-ketoglutarate from the TCA cycle to the syntheses of amino acids, such as arginine and leucine. This mitochondrial response achieves a trade-off between energy metabolism and nitrogenic anabolism, which serves as an effector mechanism promoting cell survival under MR.
Collapse
Affiliation(s)
- Wen Fang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Liu Jiang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yibing Zhu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Sen Yang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Hong Qiu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Jiou Cheng
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Qingxi Liang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, 330022, China
| | - Zong-Cai Tu
- National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, 330022, China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Cunqi Ye
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
48
|
Siani G, Mercaldo B, Alterisio MC, Di Loria A. Vitamin B12 in Cats: Nutrition, Metabolism, and Disease. Animals (Basel) 2023; 13:ani13091474. [PMID: 37174511 PMCID: PMC10177498 DOI: 10.3390/ani13091474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Cobalamin is a water-soluble molecule that has an important role in cellular metabolism, especially in DNA synthesis, methylation, and mitochondrial metabolism. Cobalamin is bound by intrinsic factor (IF) and absorbed in the ileal tract. The IF in cats is synthesized exclusively by pancreatic tissue. About 75% of the total plasma cobalamin in cats is associated with transcobalamin II, while in this species, transcobalamin I is not present. In cats, the half-life of cobalamin is 11-14 days. Diagnostic biomarkers for B12 status in cats include decreased levels of circulating total cobalamin and increased levels of methylmalonic acid. The reference interval for serum cobalamin concentrations in cats is 290-1500 ng/L, and for the serum methylmalonic acid concentration, it is 139-897 nmol/L. Therapy for hypocobalaminemia mainly depends on the underlying disease. In some cases, subcutaneous or intramuscular injection of 250 μg/cat is empirically administered. In recent years, it has been demonstrated that oral cobalamin supplementation can also be used successfully in dogs and cats as a less invasive alternative to parental administration. This review describes the current knowledge regarding B12 requirements and highlights improvements in diagnostic methods as well as the role of hypocobalaminemia in its associated diseases.
Collapse
Affiliation(s)
| | - Beatrice Mercaldo
- Department of Veterinary Medicine and Animal Productions, University Federico II of Napoli, Via F. Delpino 1, 80137 Napoli, Italy
| | - Maria Chiara Alterisio
- Department of Veterinary Medicine and Animal Productions, University Federico II of Napoli, Via F. Delpino 1, 80137 Napoli, Italy
| | - Antonio Di Loria
- Department of Veterinary Medicine and Animal Productions, University Federico II of Napoli, Via F. Delpino 1, 80137 Napoli, Italy
| |
Collapse
|
49
|
Sadeeshkumar H, Balaji A, Sutherland AG, Mootien S, Anthony KG, Breaker RR. Screening for small molecule inhibitors of SAH nucleosidase using an SAH riboswitch. Anal Biochem 2023; 666:115047. [PMID: 36682579 PMCID: PMC11149561 DOI: 10.1016/j.ab.2023.115047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/21/2023]
Abstract
Due to the emergence of multidrug resistant pathogens, it is imperative to identify new targets for antibiotic drug discovery. The S-adenosylhomocysteine (SAH) nucleosidase enzyme is a promising target for antimicrobial drug development due to its critical functions in multiple bacterial processes including recycling of toxic byproducts of S-adenosylmethionine (SAM)-mediated reactions and producing the precursor of the universal quorum sensing signal, autoinducer-2 (AI-2). Riboswitches are structured RNA elements typically used by bacteria to precisely monitor and respond to changes in essential bacterial processes, including metabolism. Natural riboswitches fused to a reporter gene can be exploited to detect changes in metabolism or in physiological signaling. We performed a high-throughput screen (HTS) using an SAH-riboswitch controlled β-galactosidase reporter gene in Escherichia coli to discover small molecules that inhibit SAH recycling. We demonstrate that the assay strategy using SAH riboswitches to detect the effects of SAH nucleosidase inhibitors can quickly identify compounds that penetrate the barriers of Gram-negative bacterial cells and perturb pathways involving SAH.
Collapse
Affiliation(s)
- Harini Sadeeshkumar
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, 06520-8103, USA
| | - Aparaajita Balaji
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, 06520-8103, USA
| | | | | | - Karen G Anthony
- L2 Diagnostics, LLC, 300 George Street, New Haven, CT, 06511, USA
| | - Ronald R Breaker
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, 06520-8103, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520-8103, USA; Howard Hughes Medical Institute, Yale University, New Haven, CT, 06520-8103, USA.
| |
Collapse
|
50
|
Roth C, Kilpinen H, Kurian MA, Barral S. Histone lysine methyltransferase-related neurodevelopmental disorders: current knowledge and saRNA future therapies. Front Cell Dev Biol 2023; 11:1090046. [PMID: 36923252 PMCID: PMC10009263 DOI: 10.3389/fcell.2023.1090046] [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: 12/07/2022] [Accepted: 02/06/2023] [Indexed: 03/02/2023] Open
Abstract
Neurodevelopmental disorders encompass a group of debilitating diseases presenting with motor and cognitive dysfunction, with variable age of onset and disease severity. Advances in genetic diagnostic tools have facilitated the identification of several monogenic chromatin remodeling diseases that cause Neurodevelopmental disorders. Chromatin remodelers play a key role in the neuro-epigenetic landscape and regulation of brain development; it is therefore not surprising that mutations, leading to loss of protein function, result in aberrant neurodevelopment. Heterozygous, usually de novo mutations in histone lysine methyltransferases have been described in patients leading to haploinsufficiency, dysregulated protein levels and impaired protein function. Studies in animal models and patient-derived cell lines, have highlighted the role of histone lysine methyltransferases in the regulation of cell self-renewal, cell fate specification and apoptosis. To date, in depth studies of histone lysine methyltransferases in oncology have provided strong evidence of histone lysine methyltransferase dysregulation as a determinant of cancer progression and drug resistance. As a result, histone lysine methyltransferases have become an important therapeutic target for the treatment of different cancer forms. Despite recent advances, we still lack knowledge about the role of histone lysine methyltransferases in neuronal development. This has hampered both the study and development of precision therapies for histone lysine methyltransferases-related Neurodevelopmental disorders. In this review, we will discuss the current knowledge of the role of histone lysine methyltransferases in neuronal development and disease progression. We will also discuss how RNA-based technologies using small-activating RNAs could potentially provide a novel therapeutic approach for the future treatment of histone lysine methyltransferase haploinsufficiency in these Neurodevelopmental disorders, and how they could be first tested in state-of-the-art patient-derived neuronal models.
Collapse
Affiliation(s)
- Charlotte Roth
- Molecular Neurosciences, Developmental Neurosciences Programme, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Helena Kilpinen
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Manju A. Kurian
- Molecular Neurosciences, Developmental Neurosciences Programme, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Neurology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Serena Barral
- Molecular Neurosciences, Developmental Neurosciences Programme, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|