1
|
Li B, Zhang X, Zhang Q, Zheng T, Li Q, Yang S, Shao J, Guan W, Zhang S. Nutritional strategies to reduce intestinal cell apoptosis by alleviating oxidative stress. Nutr Rev 2025; 83:e518-e532. [PMID: 38626282 DOI: 10.1093/nutrit/nuae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024] Open
Abstract
The gut barrier is the first line of defense against harmful substances and pathogens in the intestinal tract. The balance of proliferation and apoptosis of intestinal epithelial cells (IECs) is crucial for maintaining the integrity of the intestinal mucosa and its function. However, oxidative stress and inflammation can cause DNA damage and abnormal apoptosis of the IECs, leading to the disruption of the intestinal epithelial barrier. This, in turn, can directly or indirectly cause various acute and chronic intestinal diseases. In recent years, there has been a growing understanding of the vital role of dietary ingredients in gut health. Studies have shown that certain amino acids, fibers, vitamins, and polyphenols in the diet can protect IECs from excessive apoptosis caused by oxidative stress, and limit intestinal inflammation. This review aims to describe the molecular mechanism of apoptosis and its relationship with intestinal function, and to discuss the modulation of IECs' physiological function, the intestinal epithelial barrier, and gut health by various nutrients. The findings of this review may provide a theoretical basis for the use of nutritional interventions in clinical intestinal disease research and animal production, ultimately leading to improved human and animal intestinal health.
Collapse
Affiliation(s)
- Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaoli Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiayuan Shao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
2
|
Guo ZX, Ma JL, Zhang JQ, Yan LL, Zhou Y, Mao XL, Li SW, Zhou XB. Metabolic reprogramming and immunological changes in the microenvironment of esophageal cancer: future directions and prospects. Front Immunol 2025; 16:1524801. [PMID: 39925801 PMCID: PMC11802498 DOI: 10.3389/fimmu.2025.1524801] [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: 11/08/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Background Esophageal cancer (EC) is the seventh-most prevalent cancer worldwide and is a significant contributor to cancer-related mortality. Metabolic reprogramming in tumors frequently coincides with aberrant immune function alterations, and extensive research has demonstrated that perturbations in energy metabolism within the tumor microenvironment influence the occurrence and progression of esophageal cancer. Current treatment modalities for esophageal cancer primarily include encompass chemotherapy and a limited array of targeted therapies, which are hampered by toxicity and drug resistance issues. Immunotherapy, particularly immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 pathway, has exhibited promising results; however, a substantial proportion of patients remain unresponsive. The optimization of these immunotherapies requires further investigation. Mounting evidence underscores the importance of modulating metabolic traits within the tumor microenvironment (TME) to augment anti-tumor immunotherapy. Methods We selected relevant studies on the metabolism of the esophageal cancer tumor microenvironment and immune cells based on our searches of MEDLINE and PubMed, focusing on screening experimental articles and reviews related to glucose metabolism, amino acid metabolism, and lipid metabolism, as well their interactions with tumor cells and immune cells, published within the last five years. We analyzed and discussed these studies, while also expressing our own insights and opinions. Results A total of 137 articles were included in the review: 21 articles focused on the tumor microenvironment of esophageal cancer, 33 delved into research related to glucose metabolism and tumor immunology, 30 introduced amino acid metabolism and immune responses, and 17 focused on the relationship between lipid metabolism in the tumor microenvironment and both tumor cells and immune cells. Conclusion This article delves into metabolic reprogramming and immune alterations within the TME of EC, systematically synthesizes the metabolic characteristics of the TME, dissects the interactions between tumor and immune cells, and consolidates and harnesses pertinent immunotherapy targets, with the goal of enhancing anti-tumor immunotherapy for esophageal cancer and thereby offering insights into the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Zhi-Xun Guo
- Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Jia-Li Ma
- Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Jin-Qiu Zhang
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Ling-Ling Yan
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Ying Zhou
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Xin-li Mao
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, Zhejiang, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Shao-Wei Li
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, Zhejiang, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Xian-Bin Zhou
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, Zhejiang, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| |
Collapse
|
3
|
Zhu W, Fu M, Li Q, Chen X, Liu Y, Li X, Luo N, Tang W, Zhang Q, Yang F, Chen Z, Zhang Y, Peng B, Zhang Q, Zhang Y, Peng X, Hu G. Amino acid metabolism-related genes as potential biomarkers and the role of MATN3 in stomach adenocarcinoma: A bioinformatics, mendelian randomization and experimental validation study. Int Immunopharmacol 2024; 143:113253. [PMID: 39353384 DOI: 10.1016/j.intimp.2024.113253] [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: 01/09/2024] [Revised: 09/11/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is a major contributor to cancer-related mortality worldwide. Alterations in amino acid metabolism, which is integral to protein synthesis, have been observed across various tumor types. However, the prognostic significance of amino acid metabolism-related genes in STAD remains underexplored. METHODS Transcriptomic gene expression and clinical data for STAD patients were obtained from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Amino acid metabolism-related gene sets were sourced from the Gene Set Enrichment Analysis (GSEA) database. A prognostic model was built using LASSO Cox regression based on the TCGA cohort and validated with GEO datasets (GSE84433, GSE84437, GSE84426). Kaplan-Meier analysis compared overall survival (OS) between high- and low-risk groups, and ROC curves assessed model accuracy. A nomogram predicted 1-, 3-, and 5-year survival. Copy number variations (CNVs) in model genes were visualized using data from the Xena platform, and mutation profiles were analyzed with "maftools" to create a waterfall plot. KEGG and GO enrichment analyses were performed to explore biological mechanisms. Immune infiltration and related functions were evaluated via ssGSEA, and Spearman correlation analyzed associations between risk scores and immune components. The TIDE database predicted immunotherapy efficacy, while FDA-approved drug sensitivity was assessed through CellMiner database. The role of MATN3 in STAD was further examined in vitro and in vivo, including amino acid-targeted metabolomic sequencing to assess its impact on metabolism. Finally, Mendelian randomization (MR) analysis evaluated the causal relationship between the model genes and gastric cancer. RESULTS In this study, we developed a prognostic risk model for STAD based on three amino acid metabolism-related genes (SERPINE1, NRP1, MATN3) using LASSO regression analysis. CNV amplification was common in SERPINE1 and NRP1, while CNV deletion frequently occurred in MATN3. STAD patients were classified into high- and low-risk groups based on the median risk score, with the high-risk group showing worse prognosis. A nomogram incorporating the risk score and clinical factors was created to estimate 1-, 3-, and 5-year survival rates. Distinct mutation profiles were observed between risk groups, with KEGG pathway analysis showing immune-related pathways enriched in the high-risk group. High-risk scores were significantly associated with the C6 (TGF-β dominant) subtype, while low-risk scores correlated with the C4 (lymphocyte-depleted) subtype. Higher risk scores also indicated increased immune infiltration, enhanced immune functions, lower tumor purity, and poorer immunotherapy response. Model genes were linked to anticancer drug sensitivity. Manipulating MATN3 expression showed that it promoted STAD cell proliferation and migration in vitro and tumor growth in vivo. Metabolomic sequencing revealed that MATN3 knockdown elevated levels of 30 amino acid metabolites, including alpha-aminobutyric acid, glycine, and aspartic acid, while reducing (S)-β-Aminoisobutyric acid and argininosuccinic acid. MR analysis found a significant causal effect of NRP1 on gastric cancer, but no causal relationship for MATN3 or SERPINE1. CONCLUSION In conclusion, the amino acid metabolism-related prognostic model shows promise as a valuable biomarker for predicting the clinical prognosis, selecting immunotherapy and drug treatment for STAD patients. Furthermore, our study has shed light on the potential value of the MATN3 as a promising strategy for combating the progression of STAD.
Collapse
Affiliation(s)
- Wenjun Zhu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Min Fu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuanhui Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoyu Li
- Department of Oncology, Hubei Cancer Hospital, Wuhan 430000, China
| | - Na Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenhua Tang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qing Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ziqi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yiling Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bi Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiang Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuanyuan Zhang
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xiaohong Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
4
|
Yu J, Yan N, Gong Z, Ma Q, Liu J, Wu X, Deng G. Mycobacterium manipulate glutaminase 1 mediated glutaminolysis to regulate macrophage autophagy for bacteria intracellular survival. Cell Signal 2024; 124:111422. [PMID: 39307377 DOI: 10.1016/j.cellsig.2024.111422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Autophagy plays a vital role in eliminating intracellular mycobacterium. It is regulated by multiple metabolic processes including glutaminolysis. Glutaminase 1 (GLS1) is the rate-limiting enzyme of glutaminolysis and has been reported to control intracellular Gln content. However, its function on regulating autophagy in mycobacterium infected macrophage is still obscure. Hence, the current study hired mycobacterium virulent strain H37Rv or attenuated strain BCG to infect macrophage and detected the changes in cell glutaminolysis. The function of GLS1 on regulating autophagy in mycobacterium infected macrophages was further investigated. The results showed that BCG infection promoted macrophage autophagy, enhanced glutaminolysis, reduced intracellular Gln content, accompanied with the up-regulation of GLS1. Conversely, H37Rv infection resulted in completely opposite effects. Meanwhile, knockdown of GLS1 increased Gln content and attenuated autophagy in BCG infected macrophages. In addition, the deprivation of Gln not only promoted the autophagy of H37Rv infected macrophages, but also abolished the effect of knockdown GLS1 on regulating BCG infection-induced mTOR activation or autophagy. To sum up, our study suggested that different virulent strains of mycobacterium infection have totally opposite effects on glutaminolysis and the expression of GLS1. Specifically, mycobacterium virulent strain reduced GLS1 expression and decreased Gln content but mycobacterium attenuated strain promoted GLS1 expression and enhanced Gln content. Furthermore, GLS1 inhibits the activation of the mTOR signaling pathway and promotes autophagy by decreasing Gln content.
Collapse
Affiliation(s)
- Jialin Yu
- School of Life Science, NingXia University, Yinchuan, NingXia, 750021, China; Key lab of ministry of education for protection and utilization of special biological resources in western China, NingXia University, Yinchuan, NingXia, 750021, China
| | - Na Yan
- School of Life Science, NingXia University, Yinchuan, NingXia, 750021, China; Key lab of ministry of education for protection and utilization of special biological resources in western China, NingXia University, Yinchuan, NingXia, 750021, China
| | - Zhaoqian Gong
- School of Life Science, NingXia University, Yinchuan, NingXia, 750021, China; Key lab of ministry of education for protection and utilization of special biological resources in western China, NingXia University, Yinchuan, NingXia, 750021, China
| | - Qinmei Ma
- School of Life Science, NingXia University, Yinchuan, NingXia, 750021, China; Key lab of ministry of education for protection and utilization of special biological resources in western China, NingXia University, Yinchuan, NingXia, 750021, China
| | - Jing Liu
- The Fourth People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Niangxia, 750021, China
| | - Xiaoling Wu
- School of Life Science, NingXia University, Yinchuan, NingXia, 750021, China; Key lab of ministry of education for protection and utilization of special biological resources in western China, NingXia University, Yinchuan, NingXia, 750021, China.
| | - Guangcun Deng
- School of Life Science, NingXia University, Yinchuan, NingXia, 750021, China; Key lab of ministry of education for protection and utilization of special biological resources in western China, NingXia University, Yinchuan, NingXia, 750021, China.
| |
Collapse
|
5
|
Bugajova M, Raudenska M, Hanelova K, Navratil J, Gumulec J, Petrlak F, Vicar T, Hrachovinova S, Masarik M, Kalfert D, Grega M, Plzak J, Betka J, Balvan J. Glutamine and serum starvation alters the ATP production, oxidative stress, and abundance of mitochondrial RNAs in extracellular vesicles produced by cancer cells. Sci Rep 2024; 14:25815. [PMID: 39468126 PMCID: PMC11519472 DOI: 10.1038/s41598-024-73943-2] [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: 04/15/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
Induction of autophagy represents an effective survival strategy for nutrient-deprived or stressed cancer cells. Autophagy contributes to the modulation of communication within the tumor microenvironment. Here, we conducted a study of the metabolic and signaling implications associated with autophagy induced by glutamine (Gln) and serum starvation and PI3K/mTOR inhibitor and autophagy inducer NVP-BEZ235 (BEZ) in the head and neck squamous cell carcinoma (HNSCC) cell line FaDu. We compared the effect of these different types of autophagy induction on ATP production, lipid peroxidation, mitophagy, RNA cargo of extracellular vesicles (EVs), and EVs-associated cytokine secretome of cancer cells. Both BEZ and starvation resulted in a decline in ATP production. Simultaneously, Gln starvation enhanced oxidative damage of cancer cells by lipid peroxidation. In starved cells, there was a discernible fragmentation of the mitochondrial network coupled with an increase in the presence of tumor susceptibility gene 101 (TSG101) on the mitochondrial membrane, indicative of the sorting of mitochondrial cargo into EVs. Consequently, the abundance of mitochondrial RNAs (mtRNAs) in EVs released by FaDu cells was enhanced. Notably, mtRNAs were also detectable in EVs isolated from the serum of both HNSCC patients and healthy controls. Starvation and BEZ reduced the production of EVs by cancer cells, yet the characteristic molecular profile of these EVs remained unchanged. We also found that alterations in the release of inflammatory cytokines constitute a principal response to autophagy induction. Importantly, the specific mechanism driving autophagy induction significantly influenced the composition of the EVs-associated cytokine secretome.
Collapse
Affiliation(s)
- Maria Bugajova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
| | - Martina Raudenska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
| | - Klara Hanelova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
| | - Jiri Navratil
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
| | - Jaromir Gumulec
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
| | - Frantisek Petrlak
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, CZ-613 00, Czech Republic
| | - Tomas Vicar
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
| | - Sarka Hrachovinova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, CZ- 625 00, Czech Republic
| | - Michal Masarik
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Vestec, CZ-252 50, Czech Republic
| | - David Kalfert
- Department of Otorhinolaryngology and Head and Neck Surgery, First Faculty of Medicine, University Hospital Motol, Charles University, V Uvalu 84, Prague, CZ-15006, Czech Republic
| | - Marek Grega
- Department of Pathology and Molecular Medicine, 2nd Faculty of Medicine, Charles University, University Hospital Motol/ V Uvalu 84, Prague 5, CZ-15006, Czech Republic
| | - Jan Plzak
- Department of Otorhinolaryngology and Head and Neck Surgery, First Faculty of Medicine, University Hospital Motol, Charles University, V Uvalu 84, Prague, CZ-15006, Czech Republic
| | - Jan Betka
- Department of Otorhinolaryngology and Head and Neck Surgery, First Faculty of Medicine, University Hospital Motol, Charles University, V Uvalu 84, Prague, CZ-15006, Czech Republic
| | - Jan Balvan
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic.
| |
Collapse
|
6
|
Nazir U, Fu Z, Zheng X, Zafar MH, Chen Y, Yang Z, Wang Z, Yang H. Effects of Alanyl-Glutamine Dipeptide Supplementation on Growth Performance, Nutrient Digestibility, Digestive Enzyme Activity, Immunity, and Antioxidant Status in Growing Laying Hens. Animals (Basel) 2024; 14:2934. [PMID: 39457865 PMCID: PMC11503830 DOI: 10.3390/ani14202934] [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: 09/04/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Alanyl-glutamine (Aln-Gln), a highly soluble and stable Glutamine-dipeptide, is known to improve the performance of poultry birds. This study aimed to investigate the effect of Aln-Gln during the rearing period on growth performance, nutrient digestibility, digestive enzyme activity, immunity, antioxidant status and relative gene expression of Hy-Line brown hens. A total of 480 healthy day-old Hy-line brown chicks with similar body weights were randomly divided into four dietary groups (8 replicates/group and 15 birds/replicate). Groups A, B, C and D were fed diets containing 0%, 0.1%, 0.2% and 0.3% Aln-Gln, respectively, for 6 weeks. The body weight (BW) and average daily gain (ADG) were higher in hens fed test diets compared with the control (p < 0.05). The feed conversion ratio (FCR) was better in test groups as compared to the control group (p < 0.05). The ADFI showed no significant difference between the groups. Dietary treatments had no effect on dry matter (DM), organic matter (OM) and crude fiber (CF) digestibility. The Aln-Gln also improved gross energy (GE) and crude protein (CP) digestibility (p < 0.05). It has also increased IgG levels in groups C and D. IgM levels were similar to the control in B, C and D. The Aln-Gln increased IL-1 in B and C, IL-2 in C and D, and IL-6 in all test groups (p < 0.05). The supplementation of Aln-Gln had no effect on serum antioxidant indices like CAT, MDA, GSH-PX, GSH, and SOD in 42-day-old growing hens. Aln-Gln supplementation had no significant effect (p > 0.05) on the activity of amylase and lipase, however, a significant improvement (p < 0.05) in the activities of trypsin and chymotrypsin was observed in the test groups. Supplemented Aln-Gln levels in the birds' diets led to an increase in the expression of genes related to growth factors (IGF-1, IGFBP-5), immune markers (IL-1, IL-2, IL-6) and antioxidant status (GSH-Px1), as compared to control group. Aln-Gln supplementation in Hy-Line brown hens during their growing period improved growth, nutrient digestibility, immunity and digestive enzymes activity. These findings suggest that Aln-Gln is a promising dietary additive for enhancing poultry performance.
Collapse
Affiliation(s)
- Usman Nazir
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zhenming Fu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Xucheng Zheng
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Muhammad Hammad Zafar
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yuanjing Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhi Yang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Zhiyue Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Haiming Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
7
|
Jin C, Zhu M, Ye J, Song Z, Zheng C, Chen W. Autophagy: Are Amino Acid Signals Dependent on the mTORC1 Pathway or Independent? Curr Issues Mol Biol 2024; 46:8780-8793. [PMID: 39194736 DOI: 10.3390/cimb46080519] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Autophagy is a kind of "self-eating" phenomenon that is ubiquitous in eukaryotic cells. It mainly manifests in the damaged proteins or organelles in the cell being wrapped and transported by the autophagosome to the lysosome for degradation. Many factors cause autophagy in cells, and the mechanism of nutrient-deficiency-induced autophagy has been a research focus. It has been reported that amino-acid-deficiency-induced cellular autophagy is mainly mediated through the mammalian rapamycin target protein complex 1 (mTORC1) signaling pathway. In addition, some researchers also found that non-mTORC1 signaling pathways also regulate autophagy, and the mechanism of autophagy occurrence induced by the deficiency of different amino acids is not precisely the same. Therefore, this review aims to summarize the process of various amino acids regulating cell autophagy and provide a narrative review on the molecular mechanism of amino acids regulating autophagy.
Collapse
Affiliation(s)
- Chenglong Jin
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Min Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Jinling Ye
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Zhiwen Song
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Chuntian Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Wei Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| |
Collapse
|
8
|
Barnaba C, Broadbent DG, Kaminsky EG, Perez GI, Schmidt JC. AMPK regulates phagophore-to-autophagosome maturation. J Cell Biol 2024; 223:e202309145. [PMID: 38775785 PMCID: PMC11110907 DOI: 10.1083/jcb.202309145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/28/2024] [Accepted: 05/04/2024] [Indexed: 05/24/2024] Open
Abstract
Autophagy is an important metabolic pathway that can non-selectively recycle cellular material or lead to targeted degradation of protein aggregates or damaged organelles. Autophagosome formation starts with autophagy factors accumulating on lipid vesicles containing ATG9. These phagophores attach to donor membranes, expand via ATG2-mediated lipid transfer, capture cargo, and mature into autophagosomes, ultimately fusing with lysosomes for their degradation. Autophagy can be activated by nutrient stress, for example, by a reduction in the cellular levels of amino acids. In contrast, how autophagy is regulated by low cellular ATP levels via the AMP-activated protein kinase (AMPK), an important therapeutic target, is less clear. Using live-cell imaging and an automated image analysis pipeline, we systematically dissect how nutrient starvation regulates autophagosome biogenesis. We demonstrate that glucose starvation downregulates autophagosome maturation by AMPK-mediated inhibition of phagophore tethering to donor membrane. Our results clarify AMPKs regulatory role in autophagy and highlight its potential as a therapeutic target to reduce autophagy.
Collapse
Affiliation(s)
- Carlo Barnaba
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - David G. Broadbent
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Emily G. Kaminsky
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Gloria I. Perez
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Jens C. Schmidt
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
9
|
Kim M, Hwang S, Jeong SM. Targeting cellular adaptive responses to glutaminolysis perturbation for cancer therapy. Mol Cells 2024; 47:100096. [PMID: 39038517 PMCID: PMC11342766 DOI: 10.1016/j.mocell.2024.100096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024] Open
Abstract
Metabolic aberrations, notably deviations in glutamine metabolism, are crucial in the oncogenic process, offering vital resources for the unlimited proliferation and enhanced survival capabilities of cancer cells. The dependency of malignant cells on glutamine metabolism has led to the proposition of targeted therapeutic strategies. However, the capability of cancer cells to initiate adaptive responses undermines the efficacy of these therapeutic interventions. This review meticulously examines the multifaceted adaptive mechanisms that cancer cells deploy to sustain survival and growth following the disruption of glutamine metabolism. Emphasis is placed on the roles of transcription factors, alterations in metabolic pathways, the mechanistic target of rapamycin complex 1 signaling axis, autophagy, macropinocytosis, nucleotide biosynthesis, and the scavenging of ROS. Thus, the delineation and subsequent targeting of these adaptive responses in the context of therapies aimed at glutamine metabolism offer a promising avenue for circumventing drug resistance in cancer treatment.
Collapse
Affiliation(s)
- Minjoong Kim
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Sunsook Hwang
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Seung Min Jeong
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea.
| |
Collapse
|
10
|
Zhou X, Liang J, Xiong X, Yin Y. Amino acids in piglet diarrhea: Effects, mechanisms and insights. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 16:267-274. [PMID: 38362520 PMCID: PMC10867606 DOI: 10.1016/j.aninu.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/28/2023] [Accepted: 07/12/2023] [Indexed: 02/17/2024]
Abstract
Piglet diarrhea is among one of the most serious health problems faced by the pig industry, resulting in significant economic losses. Diarrheal disease in piglets has a multifactorial etiology that is affected by physiology, environment, and management strategy. Diarrhea is the most apparent symptom of intestinal dysfunction. As a key class of essential nutrients in the piglet diet, amino acids confer a variety of beneficial effects on piglets in addition to being used as a substrate for protein synthesis, including maintaining appropriate intestinal integrity, permeability and epithelial renewal, and alleviating morphological damage and inflammatory and oxidative stress. Thus, provision of appropriate levels of amino acids could alleviate piglet diarrhea. Most amino acid effects are mediated by metabolites, gut microbes, and related signaling pathways. In this review, we summarize the current understanding of dietary amino acid effects on gut health and diarrhea incidence in piglets, and reveal the mechanisms involved. We also provide ideas for using amino acid blends and emphasize the importance of amino acid balance in the diet to prevent diarrhea in piglets.
Collapse
Affiliation(s)
- Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Liang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xia Xiong
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
11
|
Barnaba C, Broadbent DG, Perez GI, Schmidt JC. AMPK Regulates Phagophore-to-Autophagosome Maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.559981. [PMID: 37808644 PMCID: PMC10557706 DOI: 10.1101/2023.09.28.559981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Autophagy is an important metabolic pathway that can non-selectively recycle cellular material or lead to targeted degradation of protein aggregates or damaged organelles. Autophagosome formation starts with autophagy factors accumulating on lipid vesicles containing ATG9. These phagophores attach to donor membranes, expand via ATG2-mediated lipid transfer, capture cargo, and mature into autophagosomes, ultimately fusing with lysosomes for their degradation. Autophagy can be activated by nutrient stress, for example by a reduction in the cellular levels of amino acids. In contrast, how autophagy is regulated by low cellular ATP levels via the AMP-activated protein kinase (AMPK), an important therapeutic target, is less clear. Using live-cell imaging and an automated image analysis pipeline, we systematically dissect how nutrient starvation regulates autophagosome biogenesis. We demonstrate that glucose starvation downregulates autophagosome maturation by AMPK mediated inhibition of phagophores tethering to donor membranes. Our results clarify AMPK's regulatory role in autophagy and highlight its potential as a therapeutic target to reduce autophagy.
Collapse
Affiliation(s)
- Carlo Barnaba
- Institute for Quantitative Health Science and Engineering
| | - David G. Broadbent
- Institute for Quantitative Health Science and Engineering
- College of Osteopathic Medicine
- Department of Physiology
| | | | - Jens C. Schmidt
- Institute for Quantitative Health Science and Engineering
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, USA
| |
Collapse
|
12
|
Zhang X, Wang A, Chang E, Han B, Xu J, Fu Y, Dong X, Miao S. Effects of dietary tryptophan on the antioxidant capacity and immune response associated with TOR and TLRs/MyD88/NF-κB signaling pathways in northern snakehead, Channa argus (Cantor, 1842). Front Immunol 2023; 14:1149151. [PMID: 37114056 PMCID: PMC10128191 DOI: 10.3389/fimmu.2023.1149151] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction Dietary tryptophan (Trp) has been shown to influence fish feed intake, growth, immunity and inflammatory responses. The purpose of this study was to investigate the effect and mechanism of Trp on immune system of juvenile northern snakehead (Channa argus Cantor, 1842). Methods A total of 540 fish (10.21 ± 0.11 g) were fed six experimental diets containing graded levels of Trp at 1.9, 3.0, 3.9, 4.8, 5.9 and 6.8 g/kg diet for 70 days, respectively. Results and Discussion The results showed that supplementation of 1.9-4.8 g/kg Trp in diets had no effect on the hepatosomatic index (HSI) and renal index (RI), while dietary 3.9 and 4.8 g/kg Trp significantly increased spleen index (SI) of fish. Dietary 3.9, 4.8, 5.9 and 6.8 g/kg Trp enhanced the total hemocyte count (THC), the activities of total antioxidant capacity (T-AOC) and superoxide dismutase (SOD). Malondinaldehyde (MDA) levels in the blood were significantly decreased by consuming 3.9 and 4.8 g/kg Trp. Fish fed with 3.0 and 3.9 g/kg Trp diets up-regulated interleukin 6 (il-6) and interleukin 8 (il-8) mRNA levels. The expression of tumor necrosis factor α (tnf-α) was highest in fish fed with 3.0 g/kg Trp diet, and the expression of interleukin 1β (il-1β) was highest in fish fed with 3.9 g/kg Trp diet. Dietary 4.8, 5.9 and 6.8 g/kg Trp significantly decreased il-6 and tnf-α mRNA levels in the intestine. Moreover, Trp supplementation was also beneficial to the mRNA expression of interleukin 22 (il-22). Additionally, the mRNA expression levels of target of rapamycin (tor), toll-like receptor-2 (tlr2), toll-like receptor-4 (tlr4), toll-like receptor-5 (tlr5) and myeloid differentiation primary response 88 (myd88) of intestine were significantly up-regulated in fish fed 1.9, 3.0 and 3.9 g/kg Trp diets, and down-regulated in fish fed 4.8, 5.9 and 6.8 g/kg Trp diets. Dietary 4.8 and 5.9 g/kg Trp significantly increased the expression of inhibitor of nuclear factor kappa B kinase beta subunit (ikkβ) and decreased the expression of inhibitor of kappa B (iκbα), but inhibited nuclear transcription factor kappa B (nf-κb) mRNA level. Collectively, these results indicated that dietary 4.8 g/kg Trp could improve antioxidant capacity and alleviate intestinal inflammation associated with TOR and TLRs/MyD88/NF-κB signaling pathways.
Collapse
|
13
|
Xiong J, Luu TTT, Venkatachalam K, Du G, Zhu MX. Glutamine Produces Ammonium to Tune Lysosomal pH and Regulate Lysosomal Function. Cells 2022; 12:80. [PMID: 36611873 PMCID: PMC9819001 DOI: 10.3390/cells12010080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Glutamine is one of the most abundant amino acids in the cell. In mitochondria, glutaminases 1 and 2 (GLS1/2) hydrolyze glutamine to glutamate, which serves as the precursor of multiple metabolites. Here, we show that ammonium generated during GLS1/2-mediated glutaminolysis regulates lysosomal pH and in turn lysosomal degradation. In primary human skin fibroblasts BJ cells and mouse embryonic fibroblasts, deprivation of total amino acids for 1 h increased lysosomal degradation capacity as shown by the increased turnover of lipidated microtubule-associated proteins 1A/1B light chain 3B (LC3-II), several autophagic receptors, and endocytosed DQ-BSA. Removal of glutamine but not any other amino acids from the culture medium enhanced lysosomal degradation similarly as total amino acid starvation. The presence of glutamine in regular culture media increased lysosomal pH by >0.5 pH unit and the removal of glutamine caused lysosomal acidification. GLS1/2 knockdown, GLS1 antagonist, or ammonium scavengers reduced lysosomal pH in the presence of glutamine. The addition of glutamine or NH4Cl prevented the increase in lysosomal degradation and curtailed the extension of mTORC1 function during the early time period of amino acid starvation. Our findings suggest that glutamine tunes lysosomal pH by producing ammonium, which regulates lysosomal degradation to meet the demands of cellular activities. During the early stage of amino acid starvation, the glutamine-dependent mechanism allows more efficient use of internal reserves and endocytosed proteins to extend mTORC1 activation such that the normal anabolism is not easily interrupted by a brief disruption of the amino acid supply.
Collapse
Affiliation(s)
- Jian Xiong
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Thi Thu Trang Luu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Program in Neuroscience, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Guangwei Du
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Program in Neuroscience, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
14
|
Kaur R, Kanthaje S, Taneja S, Dhiman RK, Chakraborti A. miR-23b-3p Modulating Cytoprotective Autophagy and Glutamine Addiction in Sorafenib Resistant HepG2, a Hepatocellular Carcinoma Cell Line. Genes (Basel) 2022; 13:genes13081375. [PMID: 36011286 PMCID: PMC9407556 DOI: 10.3390/genes13081375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is the second most common malignancy with increasing cancer deaths worldwide. HCC is mainly diagnosed at its advanced stage, and treatment with FDA-approved sorafenib, the multikinase inhibitor drug, is advised. Acquired resistance against sorafenib develops through several pathways involving hypoxia, autophagy, high glycolysis, or glutaminolysis. Small non-coding RNAs, similar to microRNAs (miRNAs), are also known to affect sorafenib resistance in HCC. However, there is a lack of information regarding the significance of differentially expressed miRNA (if any) on autophagy and glutamine regulation in sorafenib-resistant HCC. Methods: The expression of autophagy and glutaminolysis genes was checked in both parental and sorafenib resistant HepG2 cell lines by real-time PCR. MTT and Annexin/PI assays were also performed in the presence of inhibitors such as chloroquine (autophagy inhibitor) and BPTES (glutaminolysis inhibitor). Next generation sequencing and in silico analysis were performed to select autophagy and glutamine addiction-specific microRNA. Selected miRNA were transfected into both HepG2 cells to examine its effect on autophagy and glutamine addiction in regulating sorafenib-resistant HCC. Results: Our in vitro study depicted a higher expression of genes encoding autophagy and glutaminolysis in sorafenib-resistant HepG2 cells. Moreover, inhibitors for autophagy (chloroquine) and glutaminolysis (BPTES) showed a diminished level of cell viability and augmentation in cell apoptosis of sorafenib-resistant HepG2 cells. NGS and real-time PCR demonstrated the downregulated expression of miR-23b-3p in sorafenib-resistant cells compared to parental cells. In silico analysis showed that miR-23b-3p specifically targeted autophagy through ATG12 and glutaminolysis through GLS1. In transfection assays, mimics of miR-23b-3p demonstrated reduced gene expression for both ATG12 and GLS1, decreased cell viability, and increased cell apoptosis of sorafenib-resistant HepG2 cells, whereas the antimiRs of miR-23b-3p demonstrated contrasting results. Conclusion: Our study highlights the cytoprotective role of autophagy and glutamine addiction modulated by miR-23b-3p (tumor suppressor), suggesting new approaches to curb sorafenib resistance in HCC.
Collapse
Affiliation(s)
- Ramanpreet Kaur
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India; (R.K.); (S.K.)
- Department of Hepatology, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India; (S.T.); (R.K.D.)
| | - Shruthi Kanthaje
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India; (R.K.); (S.K.)
| | - Sunil Taneja
- Department of Hepatology, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India; (S.T.); (R.K.D.)
| | - Radha K. Dhiman
- Department of Hepatology, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India; (S.T.); (R.K.D.)
| | - Anuradha Chakraborti
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India; (R.K.); (S.K.)
- Correspondence: ; Tex.: +91-9876163975
| |
Collapse
|
15
|
Zhou J, Chen H, Du J, Tai H, Han X, Huang N, Wang X, Gong H, Yang M, Xiao H. Glutamine Availability Regulates the Development of Aging Mediated by mTOR Signaling and Autophagy. Front Pharmacol 2022; 13:924081. [PMID: 35860029 PMCID: PMC9289448 DOI: 10.3389/fphar.2022.924081] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Glutamine is a conditionally essential amino acid involved in energy production and redox homeostasis. Aging is commonly characterized by energy generation reduction and redox homeostasis dysfunction. Various aging-related diseases have been reported to be accompanied by glutamine exhaustion. Glutamine supplementation has been used as a nutritional therapy for patients and the elderly, although the mechanism by which glutamine availability affects aging remains elusive. Here, we show that chronic glutamine deprivation induces senescence in fibroblasts and aging in Drosophila melanogaster, while glutamine supplementation protects against oxidative stress-induced cellular senescence and rescues the D-galactose-prompted progeria phenotype in mice. Intriguingly, we found that long-term glutamine deprivation activates the Akt-mTOR pathway, together with the suppression of autolysosome function. However, the inhibition of the Akt-mTOR pathway effectively rescued the autophagy impairment and cellular senescence caused by glutamine deprivation. Collectively, our study demonstrates a novel interplay between glutamine availability and the aging process. Mechanistically, long-term glutamine deprivation could evoke mammalian target of rapamycin (mTOR) pathway activation and autophagy impairment. These findings provide new insights into the connection between glutamine availability and the aging process.
Collapse
Affiliation(s)
- Jiao Zhou
- Department of Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Honghan Chen
- Department of Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jintao Du
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Haoran Tai
- Department of Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, Chengdu Medical College, Chengdu, China
| | - Xiaojuan Han
- Department of Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ning Huang
- Department of Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaobo Wang
- Department of Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Gong
- Department of Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Mingyao Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Hengyi Xiao
- Department of Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Hengyi Xiao,
| |
Collapse
|
16
|
Zhao L, Liu Y, Zhang S, Wei L, Cheng H, Wang J, Wang J. Impacts and mechanisms of metabolic reprogramming of tumor microenvironment for immunotherapy in gastric cancer. Cell Death Dis 2022; 13:378. [PMID: 35444235 PMCID: PMC9021207 DOI: 10.1038/s41419-022-04821-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/04/2022] [Indexed: 02/07/2023]
Abstract
Metabolic disorders and abnormal immune function changes occur in tumor tissues and cells to varying degrees. There is increasing evidence that reprogrammed energy metabolism contributes to the development of tumor suppressive immune microenvironment and influences the course of gastric cancer (GC). Current studies have found that tumor microenvironment (TME) also has important clinicopathological significance in predicting prognosis and therapeutic efficacy. Novel approaches targeting TME therapy, such as immune checkpoint blockade (ICB), metabolic inhibitors and key enzymes of immune metabolism, have been involved in the treatment of GC. However, the interaction between GC cells metabolism and immune metabolism and how to make better use of these immunotherapy methods in the complex TME in GC are still being explored. Here, we discuss how metabolic reprogramming of GC cells and immune cells involved in GC immune responses modulate anti-tumor immune responses, as well as the effects of gastrointestinal flora in TME and GC. It is also proposed how to enhance anti-tumor immune response by understanding the targeted metabolism of these metabolic reprogramming to provide direction for the treatment and prognosis of GC.
Collapse
Affiliation(s)
- Lin Zhao
- The First Clinical College, Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Yuanyuan Liu
- The First Clinical College, Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Simiao Zhang
- The First Clinical College, Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Lingyu Wei
- Collaborative Innovation Center for Aging Mechanism Research and Transformation, Center for Healthy Aging, Changzhi Medical College, Changzhi, Shanxi, 046000, China.,Key Laboratory of Esophageal Cancer Basic Research and Clinical Transformation, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Hongbing Cheng
- Collaborative Innovation Center for Aging Mechanism Research and Transformation, Center for Healthy Aging, Changzhi Medical College, Changzhi, Shanxi, 046000, China.,Department of Microbiology, Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Jinsheng Wang
- Collaborative Innovation Center for Aging Mechanism Research and Transformation, Center for Healthy Aging, Changzhi Medical College, Changzhi, Shanxi, 046000, China. .,Key Laboratory of Esophageal Cancer Basic Research and Clinical Transformation, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China.
| | - Jia Wang
- Collaborative Innovation Center for Aging Mechanism Research and Transformation, Center for Healthy Aging, Changzhi Medical College, Changzhi, Shanxi, 046000, China. .,Department of Immunology, Center for Healthy Aging, Changzhi Medical College, Changzhi, Shanxi, 046000, China.
| |
Collapse
|
17
|
Wang J, Wang N, Qi M, Li J, Tan B. Glutamine, glutamate, and aspartate differently modulate energy homeostasis of small intestine under normal or low energy status in piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 8:216-226. [PMID: 34977390 PMCID: PMC8685906 DOI: 10.1016/j.aninu.2021.07.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/01/2021] [Accepted: 07/08/2021] [Indexed: 06/14/2023]
Abstract
Weaning stress may cause reduced energy intake for maintenance of mucosal structure. Gln, Glu, and Asp are major energy sources for the small intestine. This study investigated whether Gln, Glu, and Asp improve the intestinal morphology via regulating the energy metabolism in weaning piglets. A total of 198 weaned piglets were assigned to 3 treatments: Control (Basal diet + 1.59% L-Ala); T1 (Basal diet + 1% L-Gln + 0.5% L-Glu + 0.1% L-Asp); T2 (Low energy diet + 1% L-Gln + 0.5% L-Glu + 0.1% L-Asp). Jejunum and ileum were obtained on d 5 or 21 post-weaning. T1 enhanced growth performance. T1 and T2 treatments improved small intestinal morphology by increasing villus height, goblet cell number and decreasing crypt depth. Days post-weaning affected the efficacy of T2, but not T1, on energy metabolism. At normal energy supplementation, Gln, Glu, and Asp restored small intestinal energy homeostasis via replenishing the Krebs' cycle and down-regulating the AMPK (adenosine monophosphate activated protein kinase) pathway. As these are not sufficient to maintain the intestinal energy-balance of piglets fed with a low energy diet on d 5 post-weaning, the AMPK, glycolysis, beta-oxidation, and mitochondrial biogenesis are activated to meet the high energy demand of enterocytes. These data indicated that Gln, Glu, and Asp could restore the energy homeostasis of intestinal mucosa of weaning piglets under normal energy fed. Low energy feeding may increase the susceptibility of piglets to stress, which may decrease the efficacy of Gln, Glu, and Asp on the restoration of energy balance. These findings provide new information on nutritional intervention for insufficient energy intake in weaning piglets.
Collapse
Affiliation(s)
- Jing Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, Hunan, China
- Animal Nutrition and Human Health Laboratory, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Nan Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Ming Qi
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agroecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, Hunan, China
- University of Chinese Academy of Sciences, Beijing 10008, China
| | - Jianjun Li
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agroecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, Hunan, China
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, Hunan, China
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agroecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, Hunan, China
| |
Collapse
|
18
|
Liu WJ, Pan PY, Sun Y, Wang JB, Zhou H, Xie X, Duan ZY, Dong HY, Chen WN, Zhang LD, Wang C. Deferoxamine Counteracts Cisplatin Resistance in A549 Lung Adenocarcinoma Cells by Increasing Vulnerability to Glutamine Deprivation-Induced Cell Death. Front Oncol 2022; 11:794735. [PMID: 35127502 PMCID: PMC8810525 DOI: 10.3389/fonc.2021.794735] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/30/2021] [Indexed: 02/06/2023] Open
Abstract
Glutamine, like glucose, is a major nutrient consumed by cancer cells, yet these cells undergo glutamine starvation in the cores of tumors, forcing them to evolve adaptive metabolic responses. Pharmacologically targeting glutamine metabolism or withdrawal has been exploited for therapeutic purposes, but does not always induce cancer cell death. The mechanism by which cancer cells adapt to resist glutamine starvation in cisplatin-resistant non-small-cell lung cancer (NSCLC) also remains uncertain. Here, we report the potential metabolic vulnerabilities of A549/DDP (drug-resistant human lung adenocarcinoma cell lines) cells, which were more easily killed by the iron chelator deferoxamine (DFO) during glutamine deprivation than their parental cisplatin-sensitive A549 cells. We demonstrate that phenotype resistance to cisplatin is accompanied by adaptive responses during glutamine deprivation partly via higher levels of autophagic activity and apoptosis resistance characteristics. Moreover, this adaptation could be explained by sustained glucose instead of glutamine-dominant complex II-dependent oxidative phosphorylation (OXPHOS). Further investigation revealed that cisplatin-resistant cells sustain OXPHOS partly via iron metabolism reprogramming during glutamine deprivation. This reprogramming might be responsible for mitochondrial iron-sulfur [Fe-S] cluster biogenesis, which has become an “Achilles’ heel,” rendering cancer cells vulnerable to DFO-induced autophagic cell death and apoptosis through c-Jun N-terminal kinase (JNK) signaling. Finally, in vivo studies using xenograft mouse models also confirmed the growth-slowing effect of DFO. In summary, we have elucidated the adaptive responses of cisplatin-resistant NSCLC cells, which balanced stability and plasticity to overcome metabolic reprogramming and permitted them to survive under stress induced by chemotherapy or glutamine starvation. In addition, for the first time, we show that suppressing the growth of cisplatin-resistant NSCLC cells via iron chelator-induced autophagic cell death and apoptosis was possible with DFO treatment. These findings provide a solid basis for targeting mitochondria iron metabolism in cisplatin-resistant NSCLC for therapeutic purposes, and it is plausible to consider that DFO facilitates in the improvement of treatment responses in cisplatin-resistant NSCLC patients.
Collapse
Affiliation(s)
- Wen-Jun Liu
- Teaching and Experimental Center, Liaoning University of Traditional Chinese Medicine, Shenyang, China.,Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Peng-Yu Pan
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Ye Sun
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Shenyang, China
| | - Jian-Bo Wang
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine (TCM) Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Huan Zhou
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xin Xie
- Teaching and Experimental Center, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Zhi-Yuan Duan
- Teaching and Experimental Center, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Han-Yu Dong
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Wen-Na Chen
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine (TCM) Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Li-de Zhang
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Chun Wang
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
19
|
Fares HM, Lyu X, Xu X, Dong R, Ding M, Mi S, Wang Y, Li X, Yuan S, Sun L. Autophagy in cancer: The cornerstone during glutamine deprivation. Eur J Pharmacol 2022; 916:174723. [PMID: 34973953 DOI: 10.1016/j.ejphar.2021.174723] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 12/19/2022]
Abstract
Over the past two decades, researchers have revealed the crucial functions of glutamine in supporting the hyperproliferation state of cancer cells. Glutamine acts on maintaining high energy production, supporting redox status and amino acid homeostasis. Therefore, cancer cells exhibit excessive uptake of the extracellular glutamine, synthesize it in some cases, and recycle intracellular and extracellular proteins to provide an additional source of glutamine to satisfy the increasing glutamine demand. On the other hand, autophagy's role is still debated regarding tumor initiation and progression. However, most cancer cells urgently need autophagy to overcome the existential threats during glutamine restriction stress. Downstream to various stress pathways induced during such a condition, autophagy is considered an indispensable cytoprotective tool to maintain cell integrity and survival. However, the overactivation of the autophagy process is related to lethal consequences. This review summarized glutamine pathways to control autophagy and highlighted autophagy's primary activation pathways, and discussed the roles during glutamine deprivation.
Collapse
Affiliation(s)
- Hamza M Fares
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiaodan Lyu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiaoting Xu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Renchao Dong
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Muyao Ding
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Shichao Mi
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Yifan Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xue Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Shengtao Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Li Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
20
|
Jiao C, Zou J, Chen Z, Zheng F, Xu Z, Lin YH, Wang Q. Dietary Glutamine Inclusion Regulates Immune and Antioxidant System, as Well as Programmed Cell Death in Fish to Protect against Flavobacterium columnare Infection. Antioxidants (Basel) 2021; 11:44. [PMID: 35052548 PMCID: PMC8773122 DOI: 10.3390/antiox11010044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 01/07/2023] Open
Abstract
The susceptibility of animals to pathogenic infection is significantly affected by nutritional status. The present study took yellow catfish (Pelteobagrus fulvidraco) as a model to test the hypothesis that the protective roles of glutamine during bacterial infection are largely related to its regulation on the immune and antioxidant system, apoptosis and autophagy. Dietary glutamine supplementation significantly improved fish growth performance and feed utilization. After a challenge with Flavobacterium columnare, glutamine supplementation promoted il-8 and il-1β expression via NF-κB signaling in the head kidney and spleen, but inhibited the over-inflammation in the gut and gills. Additionally, dietary glutamine inclusion also enhanced the systematic antioxidant capacity. Histological analysis showed the protective role of glutamine in gill structures. Further study indicated that glutamine alleviated apoptosis during bacterial infection, along with the reduced protein levels of caspase-3 and the reduced expression of apoptosis-related genes. Moreover, glutamine also showed an inhibitory role in autophagy which was due to the increased activation of the mTOR signaling pathway. Thus, our study for the first time illustrated the regulatory roles of glutamine in the fish immune and antioxidant system, and reported its inhibitory effects on fish apoptosis and autophagy during bacterial infection.
Collapse
Affiliation(s)
- Congrui Jiao
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| | - Jiahong Zou
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| | - Zhenwei Chen
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| | - Feifei Zheng
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| | - Zhen Xu
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| | - Yu-Hung Lin
- Department of Aquaculture, National Pingtung University of Science and Technology, 1 Shuefu Road, Neipu, Pingtung 912, Taiwan
| | - Qingchao Wang
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| |
Collapse
|
21
|
NO, way to go: critical amino acids to replenish nitric oxide production in treating mucositis. Curr Opin Support Palliat Care 2021; 15:188-196. [PMID: 34397582 DOI: 10.1097/spc.0000000000000560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW There is still an unmet need for preventive and treatment strategies for chemotherapy-induced and radiotherapy-induced mucositis and its associated systemic inflammatory response (SIR) in cancer patients. Because of citrulline depletion due to cytotoxic therapy, nitric oxide (NO) production can be reduced, limiting its effect in many physiological processes. Restoring NO production could relieve mucositis severity by supporting host damage control mechanisms. Amino acids glutamine, arginine and citrulline are involved in NO production. This review including recent literature of preclinical and clinical studies will discuss the potential benefits of glutamine, arginine and citrulline on mucositis development with focus on NO production. RECENT FINDINGS Mucositis severity is more defined by host response to DNA damage than by DMA damage itself. Citrulline depletion because of afunctional enterocytes could be responsible for NO depletion during cytotoxic therapy. Restoring NO production during cytotoxic therapy could have a beneficial effect on mucositis development. Citrulline seems a more promising NO donor than glutamine or arginine during cytotoxic therapy, although clinical studies in mucositis patients are currently lacking. SUMMARY Glutamine, arginine and citrulline show in-vitro beneficial effects on inflammatory processes involved in mucositis. Translation to the clinic is difficult as demonstrated with use of glutamine and arginine. Citrulline, being the most potent NO donor with excellent oral bio-availability, is very promising as treatment choice for mucositis and its use deserves to be investigated in clinical trials with mucositis patients.
Collapse
|
22
|
Gu M, Pan S, Li Q, Qi Z, Deng W, Bai N. Protective effects of glutamine against soy saponins-induced enteritis, tight junction disruption, oxidative damage and autophagy in the intestine of Scophthalmus maximus L. FISH & SHELLFISH IMMUNOLOGY 2021; 114:49-57. [PMID: 33887442 DOI: 10.1016/j.fsi.2021.04.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 06/12/2023]
Abstract
Soy saponins, as thermo-stable anti-nutrients in soybean meal (SBM), are the primary causal agents of SBM-induced enteritis, which represents a well-documented pathologic alternation involving the distal intestines of various farmed fish. Our previous work showed that soy saponins might lead to SBM-induced enteritis, destroy tight junction structure and induce oxidative damage in juvenile turbot. Glutamine, as a conditionally essential amino acid, is an important substrate utilized for the growth of intestinal epithelial cells. An 8-week feeding trial was carried out to determine whether glutamine can attenuate the detrimental effects of soy saponins. Three isonitrogenous-isolipidic experimental diets were formulated as follows: (i) fish meal-based diet (FM), considered as control; (ii) FM + 10 g/kg soy saponins, SAP; and (iii) SAP + 15 g/kg glutamine, GLN. The results showed that dietary soy saponins significantly increased the gene expression levels of inflammatory markers (IL-1β, IL-8 and TNF-α) and related signaling factors (NF-кB, AP-1, p38, JNK and ERK), which were remarkably attenuated by dietary glutamine. Compared to SAP group, GLN-fed fish exhibited significantly higher expression levels of tight junction genes (CLDN3, CLDN4, OCLN, Tricellulin and ZO-1). Glutamine supplementation in SAP diet markedly suppressed the production of reactive oxygen species, malondialdehyde and protein carbonyl, and enhanced the activities of antioxidant enzymes as well as the mRNA levels of HO-1, SOD, GPX and Nrf2. Furthermore, GLN-fed fish had a remarkably lower number of autophagosomes compared to SAP-fed fish. In conclusion, our study indicated that glutamine could reverse the harmful effects of soy saponins on intestinal inflammation, tight junction disruption and oxidative damage, via attenuation of NF-кB, AP-1 and MAPK pathways and activation of Nrf2 pathway. Glutamine may have the function of controlling autophaghic process within an appropriate level of encountering inflammation.
Collapse
Affiliation(s)
- Min Gu
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Shihui Pan
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Qing Li
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Zezheng Qi
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Wanzhen Deng
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Nan Bai
- Marine College, Shandong University, Weihai, Shandong, 264209, China.
| |
Collapse
|
23
|
Li M, Thorne RF, Shi R, Zhang XD, Li J, Li J, Zhang Q, Wu M, Liu L. DDIT3 Directs a Dual Mechanism to Balance Glycolysis and Oxidative Phosphorylation during Glutamine Deprivation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2003732. [PMID: 34105294 PMCID: PMC8188220 DOI: 10.1002/advs.202003732] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/29/2021] [Indexed: 05/26/2023]
Abstract
Extracellular glutamine represents an important energy source for many cancer cells and its metabolism is intimately involved in maintaining redox homeostasis. The heightened metabolic activity within tumor tissues can result in glutamine deficiency, necessitating metabolic reprogramming responses. Here, dual mechanisms involving the stress-responsive transcription factor DDIT3 (DNA damage induced transcript 3) that establishes an interrelationship between glycolysis and mitochondrial respiration are revealed. DDIT3 is induced during glutamine deprivation to promote glycolysis and adenosine triphosphate production via suppression of the negative glycolytic regulator TIGAR. In concert, a proportion of the DDIT3 pool translocates to the mitochondria and suppresses oxidative phosphorylation through LONP1-mediated down-regulation of COQ9 and COX4. This in turn dampens the sustained levels of reactive oxygen species that follow glutamine withdrawal. Together these mechanisms constitute an adaptive survival mechanism permitting tumor cells to survive metabolic stress induced by glutamine starvation.
Collapse
Affiliation(s)
- Mingyue Li
- Heifei National Laboratory for Physical Sciences at the Microscale of USTCCAS Centre for Excellence in Molecular Cell Sciencethe First Affiliated Hospital of University of Science and Technology of ChinaHefeiAnhui230027China
| | - Rick Francis Thorne
- Translational Research InstituteHenan Provincial People's HospitalSchool of Clinical MedicineHenan UniversityZhengzhouHenan450003China
| | - Ronghua Shi
- Heifei National Laboratory for Physical Sciences at the Microscale of USTCCAS Centre for Excellence in Molecular Cell Sciencethe First Affiliated Hospital of University of Science and Technology of ChinaHefeiAnhui230027China
| | - Xu Dong Zhang
- Translational Research InstituteHenan Provincial People's HospitalSchool of Clinical MedicineHenan UniversityZhengzhouHenan450003China
| | - Jingmin Li
- Translational Research InstituteHenan Provincial People's HospitalSchool of Clinical MedicineHenan UniversityZhengzhouHenan450003China
- Harbin Medical University Cancer HospitalHarbinHeilongjiang150081China
| | - Jingtong Li
- Harbin Medical University Cancer HospitalHarbinHeilongjiang150081China
| | - Qingyuan Zhang
- Harbin Medical University Cancer HospitalHarbinHeilongjiang150081China
| | - Mian Wu
- Heifei National Laboratory for Physical Sciences at the Microscale of USTCCAS Centre for Excellence in Molecular Cell Sciencethe First Affiliated Hospital of University of Science and Technology of ChinaHefeiAnhui230027China
- Translational Research InstituteHenan Provincial People's HospitalSchool of Clinical MedicineHenan UniversityZhengzhouHenan450003China
| | - Lianxin Liu
- Heifei National Laboratory for Physical Sciences at the Microscale of USTCCAS Centre for Excellence in Molecular Cell Sciencethe First Affiliated Hospital of University of Science and Technology of ChinaHefeiAnhui230027China
| |
Collapse
|
24
|
Chen S, Jiang J, Chao G, Hong X, Cao H, Zhang S. Pure Total Flavonoids From Citrus Protect Against Nonsteroidal Anti-inflammatory Drug-Induced Small Intestine Injury by Promoting Autophagy in vivo and in vitro. Front Pharmacol 2021; 12:622744. [PMID: 33953669 PMCID: PMC8090934 DOI: 10.3389/fphar.2021.622744] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
Small intestine injury is an adverse effect of non-steroidal anti-inflammatory drugs (NSAIDs) that urgently needs to be addressed for their safe application. Although pure total flavonoids from citrus (PTFC) have been marketed for the treatment of digestive diseases, their effects on small intestine injury and the underlying mechanism of action remain unknown. This study aimed to investigate the potential role of autophagy in the mechanism of NSAID (diclofenac)-induced intestinal injury in vivo and in vitro and to demonstrate the protective effects of PTFC against NSAID-induced small intestine disease. The results of qRT-PCR, western blotting, and immunohistochemistry showed that the expression levels of autophagy-related 5 (Atg5), light chain 3 (LC3)-II, and tight junction (TJ) proteins ZO-1, claudin-1, and occludin were decreased in rats with NSAID-induced small intestine injury and diclofenac-treated IEC-6 cells compared with the control groups. In the PTFC group, Atg5 and LC3-II expression, TJ protein expression, and the LC3-II/LC3-I ratio increased. Furthermore, the mechanism by which PTFC promotes autophagy in vivo and in vitro was evaluated by western blotting. Expression levels of p-PI3K and p-Akt increased in the intestine disease-induced rat model group compared with the control, but decreased in the PTFC group. Autophagy of IEC-6 cells was upregulated after treatment with a PI3K inhibitor, and the upregulation was significantly more after PTFC treatment, suggesting PTFC promoted autophagy through the PI3K/Akt signaling pathway. In conclusion, PTFC protected intestinal barrier integrity by promoting autophagy, which demonstrates its potential as a therapeutic candidate for NSAID-induced small intestine injury.
Collapse
Affiliation(s)
- Shanshan Chen
- First Affiliated Hospital, Zhejiang Chinese Medical University, Zhejiang, China
| | - Jianping Jiang
- Department of Pharmacy, School of Medicine, Zhejiang University City College, Zhejiang, China.,Zhejiang You-du Biotech Limited Company, Quzhou, China
| | | | - Xiaojie Hong
- First Affiliated Hospital, Zhejiang Chinese Medical University, Zhejiang, China
| | - Haijun Cao
- First Affiliated Hospital, Zhejiang Chinese Medical University, Zhejiang, China
| | - Shuo Zhang
- First Affiliated Hospital, Zhejiang Chinese Medical University, Zhejiang, China
| |
Collapse
|
25
|
Mitra D, Vega-Rubin-de-Celis S, Royla N, Bernhardt S, Wilhelm H, Tarade N, Poschet G, Buettner M, Binenbaum I, Borgoni S, Vetter M, Kantelhardt EJ, Thomssen C, Chatziioannou A, Hell R, Kempa S, Müller-Decker K, Wiemann S. Abrogating GPT2 in triple-negative breast cancer inhibits tumor growth and promotes autophagy. Int J Cancer 2021; 148:1993-2009. [PMID: 33368291 DOI: 10.1002/ijc.33456] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 11/09/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022]
Abstract
Uncontrolled proliferation and altered metabolic reprogramming are hallmarks of cancer. Active glycolysis and glutaminolysis are characteristic features of these hallmarks and required for tumorigenesis. A fine balance between cancer metabolism and autophagy is a prerequisite of homeostasis within cancer cells. Here we show that glutamate pyruvate transaminase 2 (GPT2), which serves as a pivot between glycolysis and glutaminolysis, is highly upregulated in aggressive breast cancers, particularly the triple-negative breast cancer subtype. Abrogation of this enzyme results in decreased tricarboxylic acid cycle intermediates, which promotes the rewiring of glucose carbon atoms and alterations in nutrient levels. Concordantly, loss of GPT2 results in an impairment of mechanistic target of rapamycin complex 1 activity as well as the induction of autophagy. Furthermore, in vivo xenograft studies have shown that autophagy induction correlates with decreased tumor growth and that markers of induced autophagy correlate with low GPT2 levels in patient samples. Taken together, these findings indicate that cancer cells have a close network between metabolic and nutrient sensing pathways necessary to sustain tumorigenesis and that aminotransferase reactions play an important role in maintaining this balance.
Collapse
Affiliation(s)
- Devina Mitra
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Silvia Vega-Rubin-de-Celis
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Nadine Royla
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Stephan Bernhardt
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Heike Wilhelm
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nooraldeen Tarade
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Gernot Poschet
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Michael Buettner
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Ilona Binenbaum
- Department of Biology, University of Patras, Patras, Greece
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
- Division of Medical Informatics for Translational Oncology, German Cancer Research Centre, Heidelberg, Germany
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens, Greece
| | - Simone Borgoni
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Martina Vetter
- Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Eva Johanna Kantelhardt
- Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Christoph Thomssen
- Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Aristotelis Chatziioannou
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
- e-NIOS PC, Athens, Greece
| | - Rüdiger Hell
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Stefan Kempa
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute of Health (BIH), Berlin, Germany
| | - Karin Müller-Decker
- DKFZ Tumor Models Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
26
|
Zhang Q, Hou Y, Bazer FW, He W, Posey EA, Wu G. Amino Acids in Swine Nutrition and Production. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1285:81-107. [PMID: 33770404 DOI: 10.1007/978-3-030-54462-1_6] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Amino acids are the building blocks of proteins in animals, including swine. With the development of new analytical methods and biochemical research, there is a growing interest in fundamental and applied studies to reexamine the roles and usage of amino acids (AAs) in swine production. In animal nutrition, AAs have been traditionally classified as nutritionally essential (EAAs) or nutritionally nonessential (NEAAs). AAs that are not synthesized de novo must be provided in diets. However, NEAAs synthesized by cells of animals are more abundant than EAAs in the body, but are not synthesized de novo in sufficient amounts for the maximal productivity or optimal health (including resistance to infectious diseases) of swine. This underscores the conceptual limitations of NEAAs in swine protein nutrition. Notably, the National Research Council (NRC 2012) has recognized both arginine and glutamine as conditionally essential AAs for pigs to improve their growth, development, reproduction, and lactation. Results of recent work have also provided compelling evidence for the nutritional essentiality of glutamate, glycine, and proline for young pigs. The inclusion of so-called NEAAs in diets can help balance AAs in diets, reduce the dietary levels of EAAs, and protect the small intestine from oxidative stress, while enhancing the growth performance, feed efficiency, and health of pigs. Thus, both EAAs and NEAAs are needed in diets to meet the requirements of pigs. This notion represents a new paradigm shift in our understanding of swine protein nutrition and is transforming pork production worldwide.
Collapse
Affiliation(s)
- Qian Zhang
- Hubei International Scientific and Technological Cooperation Base of Animal Nutrition and Gut Health, Wuhan Polytechnic University, Wuhan, China
| | - Yongqing Hou
- Hubei International Scientific and Technological Cooperation Base of Animal Nutrition and Gut Health, Wuhan Polytechnic University, Wuhan, China.
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Wenliang He
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Erin A Posey
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
27
|
Co-Chaperone Bag-1 Plays a Role in the Autophagy-Dependent Cell Survival through Beclin 1 Interaction. Molecules 2021; 26:molecules26040854. [PMID: 33561998 PMCID: PMC7914623 DOI: 10.3390/molecules26040854] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 02/06/2023] Open
Abstract
Expression levels of the major mammalian autophagy regulator Beclin 1 and its interaction with Bcl-2 regulate the switch between autophagic cell survival and apoptotic cell death pathways. However, some of the regulators and the precise mechanisms of these processes still remain elusive. Bag-1 (Bcl-2 associated athanogene-1), a member of BAG family proteins, is a multifunctional pro-survival molecule that possesses critical functions in vital cellular pathways. Herein, we report the role of Bag-1 on Bcl-2/Beclin 1 crosstalk through indirectly interacting with Beclin 1. Pull-down experiments suggested a molecular interaction between Bag-1 and Beclin 1 in breast cancer cell lines. On the other hand, in vitro binding assays showed that Bag-1/Beclin 1 interaction does not occur directly but occurs through a mediator molecule. Bag-1 interaction with p-Beclin 1 (T119), indicator of early autophagy, is increased during nutrient starvation suggesting involvement of Bag-1 in the autophagic regulation. Furthermore, CRISPR/Cas9-mediated Bag-1 knock-out in MCF-7 cells hampered cell survival and proliferation and resulted in decreased levels of total LC3 under starvation. Collectively, we suggest that Bag-1 modulates cell survival/death decision through maintaining macroautophagy as a component of Beclin 1-associated complexes.
Collapse
|
28
|
Liu T, Wang C, Wu X, Ren M, Hu Q, Jin E, Gu Y. Effect of Boron on Microstructure, Immune Function, Expression of Tight Junction Protein, Cell Proliferation and Apoptosis of Duodenum in Rats. Biol Trace Elem Res 2021; 199:205-215. [PMID: 32319072 DOI: 10.1007/s12011-020-02123-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 03/18/2020] [Indexed: 10/24/2022]
Abstract
Boron is an essential trace element for animals. Appropriate boron supplementation can produce beneficial effects on the animal body, while a high dose of boron has adverse and even toxic effects. Our aim was to investigate the impact of different doses of boron on the microstructure of duodenum in rats, expression of secretory immunoglobulin A (SIgA) and tight junction protein, cell proliferation and apoptosis. Eighty newly weaned clean Sprague-Dawley (SD) rats were given distilled water supplemented with 0, 10, 20, 40, 80, 160, 320, and 640 mg/L of boron for 60 days. We found that supplementation of 40 and 80 mg/L boron could increase the height of duodenal villi and the crypt depth, the number of intraepithelial lymphocytes (IELs) and goblet cells, the expression of SIgA, Zonula occludens-1 (ZO-1) and occludin, and proliferating cell nuclear antigen (PCNA) in duodenum of rats; decrease expression of Caspase-3 mRNA and the number of Caspase-3-positive cells, but supplementation of 320 and 640 mg/L boron could have the opposite effect in these indexes. The results showed that supplemented with 40 and 80 mg/L of boron could improve the structure and function of duodenum, while supplemented with 320-640 mg/L had a significant inhibitory effect.
Collapse
Affiliation(s)
- Ting Liu
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Chenfang Wang
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Xiaoshuang Wu
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Man Ren
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Qianqian Hu
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China.
| | - Youfang Gu
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China.
| |
Collapse
|
29
|
Ji FJ, Wang LX, Yang HS, Hu A, Yin YL. Review: The roles and functions of glutamine on intestinal health and performance of weaning pigs. Animal 2019; 13:2727-2735. [PMID: 31407650 DOI: 10.1017/s1751731119001800] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The gut is composed of a single layer of intestinal epithelial cells and plays important roles in the digestion and absorption of nutrients, immune and barrier functions and amino acid metabolism. Weaning stress impairs piglet intestinal epithelium structural and functional integrities, which results in reduced feed intake, growth rates and increased morbidity and mortality. Several measures are needed to maintain swine gut development and growth performance after weaning stress. A large body of evidence indicates that, in weaning piglets, glutamine, a functional amino acid, may improve growth performance and intestinal morphology, reduce oxidative damage, stimulate enterocyte proliferation, modulate cell survival and death and enhance intestinal paracellular permeability. This review focuses on the effects of glutamine on intestinal health in piglets. The aim is to provide evidentiary support for using glutamine as a feed additive to alleviate weaning stress.
Collapse
Affiliation(s)
- F J Ji
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, 36 Lushan Road, Changsha 410081, People's Republic of China
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, 4 West Xueyuan Road, Haikou 571101, People's Republic of China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, 644 Yuanda 2nd Road, Changsha 410125, People's Republic of China
| | - L X Wang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, 36 Lushan Road, Changsha 410081, People's Republic of China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, 644 Yuanda 2nd Road, Changsha 410125, People's Republic of China
| | - H S Yang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, 36 Lushan Road, Changsha 410081, People's Republic of China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, 644 Yuanda 2nd Road, Changsha 410125, People's Republic of China
| | - A Hu
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, 36 Lushan Road, Changsha 410081, People's Republic of China
| | - Y L Yin
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, 36 Lushan Road, Changsha 410081, People's Republic of China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, 644 Yuanda 2nd Road, Changsha 410125, People's Republic of China
- Academician Workstation of Changsha Medical University, 1501 Leifeng Road, Changsha 410219, People's Republic of China
| |
Collapse
|
30
|
Zapata RC, Singh A, Pezeshki A, Avirineni BS, Patra S, Chelikani PK. Low-Protein Diets with Fixed Carbohydrate Content Promote Hyperphagia and Sympathetically Mediated Increase in Energy Expenditure. Mol Nutr Food Res 2019; 63:e1900088. [PMID: 31365786 DOI: 10.1002/mnfr.201900088] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/17/2019] [Indexed: 12/13/2022]
Abstract
SCOPE Dietary protein restriction elicits hyperphagia and increases energy expenditure; however, less is known of whether these responses are a consequence of increasing carbohydrate content. The effects of protein-diluted diets with fixed carbohydrate content on energy balance, hormones, and key markers of protein sensing and thermogenesis in tissues are determined. METHODS AND RESULTS Obesity-prone rats (n = 13-16 per group) are randomized to diets containing fixed carbohydrate (52% calories) and varying protein concentrations: 15% (control), 10% (mild protein restriction), 5% (moderate protein restriction) or 1% (severe protein restriction) protein calories, or protein-matched to 5% protein, for 21 days. Propranolol and ondansetron are administered to interrogate the roles of sympathetic and serotonergic systems, respectively, in diet-induced changes in energy expenditure. It is found that mild-to-moderate protein restriction promotes transient hyperphagia, whereas severe protein restriction induces hypophagia, with alterations in meal patterns. Protein restriction enhances energy expenditure that is partly attenuated by propranolol, but not ondansetron. Moderate to severe protein restriction decreases gains in body weight, lean and fat mass, decreased postprandial glucose and leptin, but increased fibroblast growth factor-21 concentrations. Protein-matching retains lean mass suggesting that intake of dietary protein, but not calories, is important for preserving lean mass. Notably, protein restriction increases the protein and/or transcript abundance of key amino acid sensing molecules in liver and intestine (PERK, eIF2α, ATF2, CHOP, 4EBP1, FGF21), and upregulated thermogenic markers (β2AR, Klotho, HADH, UCP-1) in brown adipose tissue. CONCLUSION Low-protein diets promote hyperphagia and sympathetically mediated increase in energy expenditure, prevent gains in tissue reserves, and concurrently upregulate hepatic and intestinal amino acid sensing intermediaries and thermogenic markers in brown adipose tissue.
Collapse
Affiliation(s)
- Rizaldy C Zapata
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Arashdeep Singh
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Adel Pezeshki
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Bharath S Avirineni
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Souvik Patra
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Prasanth K Chelikani
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada.,Gastrointestinal Research Group, Snyder Institute for Chronic Diseases, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| |
Collapse
|
31
|
Dong X, Zhai R, Liu Z, Lin X, Wang Z, Hu Z. The Effect of Intravenous Infusions of Glutamine on Duodenal Cell Autophagy and Apoptosis in Early-Weaned Calves. Animals (Basel) 2019; 9:ani9070404. [PMID: 31266182 PMCID: PMC6680711 DOI: 10.3390/ani9070404] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/31/2019] [Accepted: 06/24/2019] [Indexed: 01/16/2023] Open
Abstract
Simple Summary The objective of this study was to determine the effects of intravenous infusions of L-glutamine (Gln) on the autophagy and apoptosis of duodenum cells in weaned calves. The results showed that the autophagy level of duodenal cells was increased with an increasing Gln infusion dose (0 to 20 g/d) and dropped when Gln was further increased to 40 g/d. We also found that the level of apoptosis was decreased with an increasing Gln infusion dose from 0 to 20 g/d, and then rose as the dose increased to 40 g/d. This knowledge will provide a reference for weaned calf health management. Abstract The objectives of this study were to determine the effects of intravenous infusions of L-glutamine (Gln) on the autophagy and apoptosis of duodenum cells in early-weaned calves. Holstein male calves were weaned at day 35 (20 male calves, birth weight 43 ± 1.8 kg; 35 ± 3 d of age) and randomly allocated to four treatments (5 calves/treatment). The treatments were: (1) infusion of NaCl, representing the control group (C); (2) infusion of 10 g/d of Gln solution (L); (3) infusion of 20 g/d of Gln solution (M); and (4) infusion of 40 g/d of Gln solution (H). The solutions were infused for 2 h daily for 3 consecutive days after weaning. All calves were killed on the third day post-weaning. The results showed that the autophagy level of the duodenal cells was increased as the Gln infusions increased from 0 to 20 g/d and dropped with a further increase in dose (40 g/d). We also found that the level of apoptosis was decreased with Gln infusion from 0 to 20 g/d and rose as the dose increased to 40 g/d. This knowledge provides a reference for weaned calf health management.
Collapse
Affiliation(s)
- Xusheng Dong
- Ruminant Nutrition and Physiology Laboratory, College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, China
| | - Ruina Zhai
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Zhaolin Liu
- Ruminant Nutrition and Physiology Laboratory, College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, China
| | - Xueyan Lin
- Ruminant Nutrition and Physiology Laboratory, College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, China
| | - Zhonghua Wang
- Ruminant Nutrition and Physiology Laboratory, College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, China
| | - Zhiyong Hu
- Ruminant Nutrition and Physiology Laboratory, College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, China.
| |
Collapse
|
32
|
Liu D, Lin J, Su J, Chen X, Jiang P, Huang K. Glutamine Deficiency Promotes PCV2 Infection through Induction of Autophagy via Activation of ROS-Mediated JAK2/STAT3 Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:11757-11766. [PMID: 30343565 DOI: 10.1021/acs.jafc.8b04704] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Porcine circovirus type 2 (PCV2) is an important pathogen in swine herds. We previously reported that glutamine (Gln) deficiency promoted PCV2 infection in vitro. Here, we established a Gln deficiency model in vivo and further investigated the detailed molecular mechanisms. In vivo and in vitro, Gln deficiency promoted PCV2 infection, which was evident through increased viral yields and PCV2 Cap protein synthesis. It also induced autophagy, as demonstrated by the increases in LC3-II conversion, SQSTM1 degradation, and GFP-LC3 dot accumulation. Autophagy inhibition abolished the effects of Gln deficiency on PCV2 infection. Inhibition of ROS generation alleviated the Gln deficiency-activated JAK2/STAT3 signaling pathway, thereby inhibiting autophagy induction. In vitro, the inhibition of STAT3 by an inhibitor or RNA interference blocked autophagy, thus reversing the effects of Gln deficiency on PCV2 infection. These results indicate that Gln deficiency activates autophagy by upregulating ROS-medicated JAK2/STAT3 signaling and thereby promoting PCV2 infection.
Collapse
|
33
|
Li J, Song P, Zhu L, Aziz N, Zhou Q, Zhang Y, Xu W, Feng L, Chen D, Wang X, Jin H. Synthetic lethality of glutaminolysis inhibition, autophagy inactivation and asparagine depletion in colon cancer. Oncotarget 2018; 8:42664-42672. [PMID: 28424408 PMCID: PMC5522096 DOI: 10.18632/oncotarget.16844] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/17/2017] [Indexed: 12/25/2022] Open
Abstract
Cancer cells reprogram metabolism to coordinate their rapid growth. They addict on glutamine metabolism for adenosine triphosphate generation and macromolecule biosynthesis. In this study, we report that glutamine deprivation retarded cell growth and induced prosurvival autophagy. Autophagy inhibition by chloroquine significantly enhanced glutamine starvation induced growth inhibition and apoptosis activation. Asparagine deprivation by L-asparaginase exacerbated growth inhibition induced by glutamine starvation and autophagy blockage. Similar to glutamine starvation, inhibition of glutamine metabolism with a chemical inhibitor currently under clinical evaluation was synthetically lethal with chloroquine and L-asparaginase, drugs approved for the treatment of malaria and leukemia, respectively. In conclusion, inhibiting glutaminolysis was synthetically lethal with autophagy inhibition and asparagine depletion. Therefore, targeting glutaminolysis could be a promising approach for colorectal cancer treatment.
Collapse
Affiliation(s)
- Jiaqiu Li
- Department of Medical Oncology, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Ping Song
- Department of Medical Oncology, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Liyuan Zhu
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Neelum Aziz
- Department of Medical Oncology, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Qiyin Zhou
- Department of Medical Oncology, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Yulong Zhang
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Wenxia Xu
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Lifeng Feng
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Dingwei Chen
- Department of Surgery, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Xian Wang
- Department of Medical Oncology, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| |
Collapse
|
34
|
Van Dyck L, Casaer MP, Gunst J. Autophagy and Its Implications Against Early Full Nutrition Support in Critical Illness. Nutr Clin Pract 2018; 33:339-347. [PMID: 29665131 DOI: 10.1002/ncp.10084] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/26/2018] [Indexed: 12/29/2022] Open
Abstract
The timing, dose, and route of early nutrition support in critically ill patients have been highly controversial for years. Despite the association of a caloric deficit with adverse outcome, several recent large, randomized, controlled trials have demonstrated a prolongation of organ failure and increased muscle weakness with increasing doses of nutrition in the acute phase of critical illness. A potential explanation for the negative impact of early, full feeding on outcome is feeding-induced suppression of autophagy, a cellular repair process that is necessary to clear intracellular damage. Whether nutrition management in critically ill patients should be guided by its effects on autophagy is a topic of debate. Currently, however, autophagy cannot be monitored in clinical practice. Moreover, clinical management should be guided by high-quality randomized controlled trials, which currently do not support the use of early full nutrition support.
Collapse
Affiliation(s)
- Lisa Van Dyck
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Belgium
| | - Michaël P Casaer
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Belgium
| | - Jan Gunst
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Belgium
| |
Collapse
|
35
|
Functions and Signaling Pathways of Amino Acids in Intestinal Inflammation. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9171905. [PMID: 29682569 PMCID: PMC5846438 DOI: 10.1155/2018/9171905] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/30/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022]
Abstract
Intestine is always exposed to external environment and intestinal microorganism; thus it is more sensitive to dysfunction and dysbiosis, leading to intestinal inflammation, such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), and diarrhea. An increasing number of studies indicate that dietary amino acids play significant roles in preventing and treating intestinal inflammation. The review aims to summarize the functions and signaling mechanisms of amino acids in intestinal inflammation. Amino acids, including essential amino acids (EAAs), conditionally essential amino acids (CEAAs), and nonessential amino acids (NEAAs), improve the functions of intestinal barrier and expressions of anti-inflammatory cytokines and tight junction proteins but decrease oxidative stress and the apoptosis of enterocytes as well as the expressions of proinflammatory cytokines in the intestinal inflammation. The functions of amino acids are associated with various signaling pathways, including mechanistic target of rapamycin (mTOR), inducible nitric oxide synthase (iNOS), calcium-sensing receptor (CaSR), nuclear factor-kappa-B (NF-κB), mitogen-activated protein kinase (MAPK), nuclear erythroid-related factor 2 (Nrf2), general controlled nonrepressed kinase 2 (GCN2), and angiotensin-converting enzyme 2 (ACE2).
Collapse
|
36
|
Csanadi A, Oser A, Aumann K, Gumpp V, Rawluk J, Nestle U, Kayser C, Wiesemann S, Werner M, Kayser G. Overexpression of SLC1a5 in lymph node metastases outperforms assessment in the primary as a negative prognosticator in non-small cell lung cancer. Pathology 2018; 50:269-275. [PMID: 29455869 DOI: 10.1016/j.pathol.2017.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 09/28/2017] [Accepted: 10/04/2017] [Indexed: 12/25/2022]
Abstract
Despite recent advances in therapeutic options, lung cancer is the leading cause of death among malignant diseases worldwide. Glutamine-dependence is an established attribute in cancer tissue with emerging importance as a diagnostic and therapeutic target. We analysed the expression of SLC1a5, a major glutamine transporter, in the primary tumour and corresponding nodal metastasis of non-small cell lung cancer (NSCLC) to investigate its biological impact. Expression of SLC1a5 was analysed by immunohistochemistry in 259 NSCLC and in 142 nodal metastases and correlated with clinicopathological parameters including overall survival. SLC1a5 expression in the primary tumour and in the corresponding lymph node metastasis revealed a positive correlation (p = 0.005). Moreover, overexpression of SLC1a5 was found to be an independent prognostic factor (p = 0.027) if assessed in lymph node metastases only. SLC1A5 expression was studied for the first time in both primary NSCLC and its corresponding nodal metastasis. Our results indicate that overexpression of SLC1a5 is associated with shorter overall survival. This proved to be an independent prognosticator if assessed in the lymph node metastases. Thus, diagnostics in lymph node metastasis provide superior prognostic information for SLC1a5 overexpression and may open target prediction for future therapeutic options.
Collapse
Affiliation(s)
- Agnes Csanadi
- Institute of Surgical Pathology, Medical Centre - University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Annika Oser
- Institute of Surgical Pathology, Medical Centre - University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Konrad Aumann
- Institute of Surgical Pathology, Medical Centre - University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Vera Gumpp
- Clinical Cancer Registry, Comprehensive Cancer Centre Freiburg, Medical Centre - University of Freiburg, Freiburg, Germany
| | - Justyna Rawluk
- Faculty of Medicine, University of Freiburg, Germany; Department of Hematology and Oncology, Medical Centre - University of Freiburg, Freiburg, Germany
| | - Ursula Nestle
- Faculty of Medicine, University of Freiburg, Germany; Department of Radiation Oncology, Medical Centre - University of Freiburg, Freiburg, Germany
| | - Claudia Kayser
- Institute of Surgical Pathology, Medical Centre - University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Sebastian Wiesemann
- Faculty of Medicine, University of Freiburg, Germany; Department of Thoracic Surgery, Medical Centre - University of Freiburg, Freiburg, Germany
| | - Martin Werner
- Institute of Surgical Pathology, Medical Centre - University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Gian Kayser
- Institute of Surgical Pathology, Medical Centre - University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
37
|
l-Glutamine Attenuates Apoptosis Induced by Endoplasmic Reticulum Stress by Activating the IRE1α-XBP1 Axis in IPEC-J2: A Novel Mechanism of l-Glutamine in Promoting Intestinal Health. Int J Mol Sci 2017; 18:ijms18122617. [PMID: 29206200 PMCID: PMC5751220 DOI: 10.3390/ijms18122617] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/28/2017] [Accepted: 12/01/2017] [Indexed: 12/18/2022] Open
Abstract
Intestinal absorption and barrier malfunctions are associated with endoplasmic reticulum stress (ERS) in the intestine. We induced ERS by exposing the intestinal porcine epithelial cell line J2 (IPEC-J2) to tunicamycin (TUNI) to explore the potential of l-glutamine to reduce ERS-induced apoptosis. Our experiments demonstrated that exposing cells to TUNI results in spontaneous ERS and encourages the upregulation of glucose-regulated protein 78 (GRP78). Prolonged TUNI-induced ERS was found to increase apoptosis mediated by C/enhancer binding protein homologous protein (CHOP), accompanied by GRP78 downregulation. Treatment with l-glutamine was found to promote cell proliferation within the growth medium but to have little effect in basic Dulbecco’s modified Eagle medium. Finally, in the milieu of TUNI-induced ERS, l-glutamine was found to maintain a high level of GRP78, alleviate CHOP-mediated apoptosis and activate the inositol requiring enzyme 1α (IRE1α)-X-box binding protein 1 (XBP1) axis. A specific inhibitor of the IRE1α-XBP1 axis reversed the protective effect of l-glutamine by blocking the expression of IRE1α/XBP1s. We propose that the functional effect of l-glutamine on intestinal health may be partly due to its modulation of ERS and CHOP-mediated apoptosis.
Collapse
|
38
|
Zhang Y, Zhao L, Zhou Y, Diao C, Han L, Yinjie N, Liu S, Chen H. Glutamine Ameliorates Mucosal Damage Caused by Immune Responses to Duck Plague Virus. Dose Response 2017; 15:1559325817708674. [PMID: 28620271 PMCID: PMC5464388 DOI: 10.1177/1559325817708674] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The immune-releasing effects of L-glutamine (Gln) supplementation in duck plague virus (DPV)-infected ducklings were evaluated in 120 seven-day-old ducklings that were divided into 8 groups. The ducklings in control and DPV, 0.5Gln and DPV + 0.5Gln, 1.0Gln and DPV + 1.0Gln, and 2.0Gln and DPV + 2.0Gln received 0, 0.5, 1.0, and 2.0 g of Gln/kg feed/d by gastric perfusion, respectively. Then, the ducklings in control to 2.0Gln were injected with 0.2 mL of phosphate-buffered saline, while those in DPV to DPV + 2.0Gln were injected with DPV at 0.2 mL of 2000 TCID50 (50% tissue culture infection dose) 30 minutes after gavage with Gln, sampled at 12 hours and days 1, 2, 4, and 6. Glutamine supplementation under physiological conditions enhanced immune function and toll-like receptor 4 (TLR4) expressions in a dose-dependent manner. An increase in Gln supplementation under DPV-infected conditions enhanced growth performance, decreased immunoglobulin (Ig) release in plasma and secretory IgA in the duodenum, ameliorated plasma cytokine levels, and suppressed overexpressions of the TLR4 pathway in the duodenum. The positive effects of Gln on the humoral immunity- and intestinal inflammation-related damage should be considered a mechanism by which immunonutrition can assist in the recovery from DPV infection.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Lili Zhao
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yan Zhou
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chenxi Diao
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Lingxia Han
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Niu Yinjie
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shengwang Liu
- Division of Avian Infectious Diseases, National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hongyan Chen
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
39
|
Zhu Y, Wang W, Yuan T, Fu L, Zhou L, Lin G, Zhao S, Zhou H, Wu G, Wang J. MicroRNA-29a mediates the impairment of intestinal epithelial integrity induced by intrauterine growth restriction in pig. Am J Physiol Gastrointest Liver Physiol 2017; 312:G434-G442. [PMID: 28280141 DOI: 10.1152/ajpgi.00020.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/22/2017] [Accepted: 03/06/2017] [Indexed: 01/31/2023]
Abstract
An important characteristic of intrauterine growth restricted (IUGR) neonate is the impaired intestinal barrier function. With the use of a pig model, this study was conducted to identify the responsible microRNA (miRNA) for the intestinal damage in IUGR neonates through comparing the miRNA profile of IUGR and normal porcine neonates and to investigate the regulation mechanism. Compared with the normal ones, we identified 83 upregulated and 76 downregulated miRNAs in the jejunum of IUGR pigs. Notably, IUGR is associated with profoundly increasesd miR-29 family and decreased expression of extracellular matrix (ECM) and tight junction (TJ) proteins in the jejunum. Furthermore, in vitro study using theporcine intestinal epithelial cell line (IPEC-1) showed that inhibition of miR-29a expression could improve the monolayer integrity by increasing cell proliferation and transepithelial resistance. Also, overexpression/inhibition of miR-29a in IPEC-1 cells can suppress/increase the expression of integrin-β1, collagen I, collagen IV, fibronectin, and claudin 1, both at transcriptional and translational levels. Subsequent luciferase reporter assay confirmed a direct interaction between miR-29a and the 3'-untranslated regions of these genes. In conclusion, this study reveals that IUGR-impaired intestinal barrier function is associated with downregulated ECM and TJ protein expression mediated by the upregulation of miR-29a.NEW & NOTEWORTHY Intrauterine growth restricted (IUGR) remains a major problem for both human health and animal production due to its association with high rates of preweaning morbidity and mortality. We have identified the abnormal expression of microRNA-29a (miR-29a) in the small intestine of IUGR neonates, as well as its targets and mechanisms. These results provide new information about biological characteristics of IUGR-affected intestinal dysfunction and can lead to the development of potentially solution for preventing and treating IUGR in the future.
Collapse
Affiliation(s)
- Yuhua Zhu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China, Agricultural University, Beijing, China
| | - Wei Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China, Agricultural University, Beijing, China
| | - Taolin Yuan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China, Agricultural University, Beijing, China
| | - Liangliang Fu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Lian Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Gang Lin
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China, Agricultural University, Beijing, China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, California; and
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China, Agricultural University, Beijing, China.,Department of Animal Science, Texas A&M University, College Station, Texas
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China, Agricultural University, Beijing, China;
| |
Collapse
|
40
|
Yi D, Hou Y, Mei H, Wang L, Hu CAA, Wu G. β-Conglycinin enhances autophagy in porcine enterocytes. Amino Acids 2017; 49:203-207. [PMID: 27761755 DOI: 10.1007/s00726-016-2352-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 10/13/2016] [Indexed: 01/09/2023]
Abstract
β-Conglycinin (β-CG) is well known for inducing intestinal allergies and dysfunction in neonates and young pigs. However, the underlying mechanisms are largely unknown. In this study, to clarify the role of autophagy in β-CG-induced cytotoxicity, we investigated the effects of β-CG on cell viability and autophagy activity in porcine enterocytes (IPEC-1 cells). The results indicated that the cell viability was decreased with the increasing levels of β-CG. β-CG treatment enhanced the eGFP-LC3 puncta per cells and LC3-II/LC3-I, and the latter was further increased in IPEC-1 cells cultured with bafilomycin A1. We conclude that β-CG enhances autophagy activity in enterocytes.
Collapse
Affiliation(s)
- Dan Yi
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yongqing Hou
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China.
| | - Huimin Mei
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Lei Wang
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Chien-An Andy Hu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Guoyao Wu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| |
Collapse
|
41
|
Autophagy is required for PDAC glutamine metabolism. Sci Rep 2016; 6:37594. [PMID: 27892481 PMCID: PMC5124864 DOI: 10.1038/srep37594] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 11/01/2016] [Indexed: 12/28/2022] Open
Abstract
Macroautophagy (autophagy) is believed to maintain energy homeostasis by degrading unnecessary cellular components and molecules. Its implication in regulating cancer metabolism recently started to be uncovered. However, the precise roles of autophagy in cancer metabolism are still unclear. Here, we show that autophagy plays a critical role in glutamine metabolism, which is required for tumor survival. Pancreatic ductal adenocarcinoma (PDAC) cells require both autophagy and typical glutamine transporters to maintain intracellular glutamine levels. Glutamine deprivation, but not that of glucose, led to the activation of macropinocytosis-associated autophagy through TFEB induction and translocation into the nucleus. In contrast, glutamine uptake increased as a compensatory response to decreased intracellular glutamine levels upon autophagy inhibition. Moreover, autophagy inhibition and glutamine deprivation did not induce cell death, while glutamine deprivation dramatically activated apoptotic cell death upon autophagy inhibition. Interestingly, the addition of α-ketoglutarate significantly rescued the apoptotic cell death caused by the combination of the inhibition of autophagy with glutamine deprivation. Our data suggest that macropinocytosis-associated autophagy is a critical process providing glutamine for anaplerosis of the TCA cycle in PDAC. Thus, targeting both autophagy and glutamine metabolism to completely block glutamine supply may provide new therapeutic approaches to treat refractory tumors.
Collapse
|
42
|
de Oliveira DC, da Silva Lima F, Sartori T, Santos ACA, Rogero MM, Fock RA. Glutamine metabolism and its effects on immune response: molecular mechanism and gene expression. ACTA ACUST UNITED AC 2016. [DOI: 10.1186/s41110-016-0016-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
43
|
Liu G, Cao W, Fang T, Jia G, Zhao H, Chen X, Wu C, Wang J. Urinary Metabolomic Approach Provides New Insights into Distinct Metabolic Profiles of Glutamine and N-Carbamylglutamate Supplementation in Rats. Nutrients 2016; 8:nu8080478. [PMID: 27527211 PMCID: PMC4997391 DOI: 10.3390/nu8080478] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 07/13/2016] [Accepted: 07/28/2016] [Indexed: 01/15/2023] Open
Abstract
Glutamine and N-carbamylglutamate can enhance growth performance and health in animals, but the underlying mechanisms are not yet elucidated. This study aimed to investigate the effect of glutamine and N-carbamylglutamate supplementation in rat metabolism. Thirty rats were fed a control, glutamine, or N-carbamylglutamate diet for four weeks. Urine samples were analyzed by nuclear magnetic resonance (NMR)-based metabolomics, specifically high-resolution 1H NMR metabolic profiling combined with multivariate data analysis. Glutamine significantly increased the urine levels of acetamide, acetate, citrulline, creatinine, and methymalonate, and decreased the urine levels of ethanol and formate (p < 0.05). Moreover, N-carbamylglutamate significantly increased the urine levels of creatinine, ethanol, indoxyl sulfate, lactate, methymalonate, acetoacetate, m-hydroxyphenylacetate, and sarcosine, and decreased the urine levels of acetamide, acetate, citrulline, creatine, glycine, hippurate, homogentisate, N-acetylglutamate, phenylacetyglycine, acetone, and p-hydroxyphenylacetate (p < 0.05). Results suggested that glutamine and N-carbamylglutamate could modify urinary metabolome related to nitrogen metabolism and gut microbiota metabolism. Moreover, N-carbamylglutamate could alter energy and lipid metabolism. These findings indicate that different arginine precursors may lead to differences in the biofluid profile in rats.
Collapse
Affiliation(s)
- Guangmang Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China.
| | - Wei Cao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China.
| | - Tingting Fang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China.
| | - Gang Jia
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China.
| | - Hua Zhao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China.
| | - Xiaoling Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China.
| | - Caimei Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China.
| | - Jing Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| |
Collapse
|
44
|
The glutamine-alpha-ketoglutarate (AKG) metabolism and its nutritional implications. Amino Acids 2016; 48:2067-80. [DOI: 10.1007/s00726-016-2254-8] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/04/2016] [Indexed: 01/08/2023]
|
45
|
Alterations of amino acid metabolism in osteoarthritis: its implications for nutrition and health. Amino Acids 2016; 48:907-914. [DOI: 10.1007/s00726-015-2168-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 12/30/2015] [Indexed: 01/15/2023]
|
46
|
Liu G, Wu X, Jia G, Zhao H, Chen X, Wu C, Wang J. Effects of glutamine against oxidative stress in the metabolome of rats—new insight. RSC Adv 2016. [DOI: 10.1039/c6ra14469a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glutamine exerts potential functions against the harmful effects of oxidative stress on animals.
Collapse
Affiliation(s)
- Guangmang Liu
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - XianJian Wu
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - Gang Jia
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - Hua Zhao
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - Xiaoling Chen
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - Caimei Wu
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - Jing Wang
- Maize Research Institute
- Sichuan Agricultural University
- Chengdu 611130
- China
| |
Collapse
|