1
|
Mandal A, Banerjee S, Ghosh S, Biswas S, Bagchi A, Sil PC. α-ketoglutarate ameliorates colitis through modulation of inflammation, ER stress, and apoptosis. Toxicol Rep 2025; 14:101897. [PMID: 39886045 PMCID: PMC11780160 DOI: 10.1016/j.toxrep.2025.101897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 02/01/2025] Open
Abstract
Colitis is an inflammatory disorder of the gastrointestinal tract. A widely consumed dietary nutrient, α-ketoglutarate (α-KG) is known to play a crucial role in cellular metabolism and provide protection to intestinal epithelium under various pathophysiological conditions. In this study, 2,4,6-trinitrobenzenesulfonic acid (TNBS) was used to induce colitis in Wistar rats. After 36 hours of TNBS administration, the rats were orally treated with a solution of α-KG at 1 g/kg body weight for 5 days. Development of colitis was confirmed by observable physical symptoms of repeated loose blood-mixed stool, apathy for food and weight loss. Macroscopic inspection revealed an inflamed colonic surface with ulcerations. Histopathological observations included alterations in crypts-structure and disruption in both epithelial and mucosal layers of colon in colitis induced rats. Colitis resulted in elevated levels of pro-inflammatory cytokines, ER stress-mediated cell death and intrinsic apoptosis pathway. The ameliorative effects of α-KG against TNBS-mediated toxicity were confirmed through molecular technics and docking analysis. Additionally, there were no instances of toxicity of α-KG. Therefore, α-KG can be considered as a valuable therapeutic agent for further comprehensive research.
Collapse
Affiliation(s)
- Ankita Mandal
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| | - Sharmistha Banerjee
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| | - Sumit Ghosh
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| | - Sima Biswas
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, Kalyani, West Bengal 741235, India
| | - Angshuman Bagchi
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, Kalyani, West Bengal 741235, India
| | - Parames C. Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| |
Collapse
|
2
|
Demianchuk O, Bayliak M, Vatashchuk M, Gospodaryov D, Hurza V, Derkachov V, Berezovskyi V, Lushchak VI. Alpha-ketoglutarate promotes anxiety, activates autophagy, and suppresses antioxidant enzymes in the cerebral cortex of female mice on cafeteria diet. Brain Res Bull 2025; 222:111255. [PMID: 39952443 DOI: 10.1016/j.brainresbull.2025.111255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/03/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025]
Abstract
Alpha-ketoglutarate (AKG), an intermediate of the tricarboxylic acid cycle, has been found to mitigate oxidative stress and inflammation. In turn, a cafeteria diet (CD), an obesogenic diet, is often associated with oxidative stress and inflammation. This study aimed to determine whether AKG can level the effects of CD on animal behavior, oxidative stress markers, glycolytic flow, and autophagy in the mouse cerebral cortex. Female C57BL/6 J mice were divided into two groups and fed either a standard diet or a CD for eight weeks. For the next four weeks, each group continued to be fed the previous diet; however, half of the individuals within each group received drinking water with 1 % AKG. Using an open field test, we found that the combination of CD and AKG promoted the development of anxiety signs. Both CD and AKG decreased the exploratory behavior of mice, with a significant additive effect in the combined diet. On diets supplemented with AKG, animals produced fewer fecal boli, a measure of emotionality. On all experimental diets, mice had lower activities of antioxidant and related enzymes, with no significant differences in the activities of glycolytic enzymes. The AKG-supplemented diet induced the transcription of autophagy-related genes and targets of the forkhead box O factor, involved in the regulation of carbohydrate metabolism. Transcriptional changes induced by AKG were partly abrogated by the CD. These findings suggest that AKG, particularly when combined with CD, may modulate behavioral responses and oxidative stress intensity in the brain by altering key metabolic and autophagic pathways.
Collapse
Affiliation(s)
- Oleh Demianchuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Maria Bayliak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine.
| | - Myroslava Vatashchuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Dmytro Gospodaryov
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine.
| | - Viktoriia Hurza
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Vitalii Derkachov
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Vladyslav Berezovskyi
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Volodymyr I Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine; Research and Development University, 13a Shota Rustaveli Str., Ivano-Frankivsk 76018, Ukraine.
| |
Collapse
|
3
|
Li M, Chen Q, Zhou M, Li X, Wang Z, Wang J. α-Ketoglutaric Acid Reprograms Macrophages by Altering Energy Metabolism to Promote the Regeneration of Small-Diameter Vascular Grafts. ACS Biomater Sci Eng 2025; 11:518-530. [PMID: 39604080 DOI: 10.1021/acsbiomaterials.4c01702] [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] [Indexed: 11/29/2024]
Abstract
Small-diameter vascular grafts still cannot clinically replace autologous blood vessels due to high restenosis rates caused by long-term inflammatory infiltration. Foreign body reactions to vascular grafts induce macrophages to adopt the pro-inflammatory M1 phenotype, releasing inflammatory factors such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). This induces a phenotypic switch in smooth muscle cells, eventually leading to intimal hyperplasia. Herein, we constructed small-diameter artificial vascular grafts capable of modulating immune responses through the controlled release of α-ketoglutaric acid (α-KG). Our findings verify that the delivery of α-KG reprograms the macrophage phenotype from a pro-inflammatory M1 to an anti-inflammatory and pro-repair M2 phenotype by regulating the energy metabolism of the tricarboxylic acid cycle (TAC). More interestingly, the delivery of α-KG positively influences the behavior of vascular cells by enhancing the proliferation of human umbilical vein endothelial cells (HUVECs) and inhibiting the expansion of mouse aortic vascular smooth muscle cells (MOVAS), thereby reducing vascular restenosis. In vivo evaluation in rabbit carotid artery replacement confirms the optimal performance of α-KG-doped vascular grafts in terms of endothelial coverage and long-term patency. Collectively, our work presents a promising approach for creating artificial vascular grafts with inflammatory regulation to ensure rapid endothelialization and sustained patency.
Collapse
Affiliation(s)
- Mengyu Li
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qi Chen
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mengxue Zhou
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaomeng Li
- National Center for International Research of Micro-Nano Molding Technology, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Zihao Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Jianglin Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
4
|
Wang Z, Hu J, Marschall JS, Yang L, Zeng E, Zhang S, Sun H. Anti-aging Metabolite-Based Polymeric Microparticles for Intracellular Drug Delivery and Bone Regeneration. SMALL SCIENCE 2024; 4:2400201. [PMID: 39386061 PMCID: PMC11460827 DOI: 10.1002/smsc.202400201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/13/2024] [Indexed: 10/12/2024] Open
Abstract
Alpha-ketoglutarate (AKG), a key component of the tricarboxylic acid (TCA) cycle, has attracted attention for its anti-aging properties. Our recent study indicates that locally delivered cell-permeable AKG significantly promotes osteogenic differentiation and mouse bone regeneration. However, the cytotoxicity and rapid hydrolysis of the metabolite limit its application. In this study, we synthesize novel AKG-based polymeric microparticles (PAKG MPs) for sustained release. In vitro data suggest that the chemical components, hydrophilicity, and size of the MPs can significantly affect their cytotoxicity and pro-osteogenic activity. Excitingly, these biodegradable PAKG MPs are highly phagocytosable for nonphagocytic pre-osteoblasts MC3T3-E1 and primary bone marrow mesenchymal stem cells (BMSCs), significantly promoting their osteoblastic differentiation. RNAseq data suggest that PAKG MPs strongly activate Wnt/β-catenin and PI3K-Akt pathways for osteogenic differentiation. Moreover, PAKG enables poly (L-lactic acid) and poly (lactic-co-glycolic acid) MPs (PLLA & PLGA MPs) for efficient phagocytosis. Our data indicate that PLGA-PAKG MPs-mediated intracellular drug delivery can significantly promote stronger osteoblastic differentiation compared to PLGA MPs-delivered phenamil. Notably, PAKG MPs significantly improve large bone regeneration in a mouse cranial bone defect model. Thus, the novel PAKG-based MPs show great promise to improve osteogenic differentiation, bone regeneration, and enable efficient intracellular drug delivery for broad regenerative medicine.
Collapse
Affiliation(s)
- Zhuozhi Wang
- Iowa Institute for Oral Health ResearchUniversity of Iowa College of DentistryIowa CityIA52242USA
| | - Jue Hu
- Iowa Institute for Oral Health ResearchUniversity of Iowa College of DentistryIowa CityIA52242USA
| | - Jeffrey S. Marschall
- Department of Oral and Maxillofacial SurgeryUniversity of Iowa College of DentistryIowa CityIA52242USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical InstituteUniversity of Iowa Carver College of MedicineIowa CityIA52242USA
| | - Erliang Zeng
- Iowa Institute for Oral Health ResearchUniversity of Iowa College of DentistryIowa CityIA52242USA
- Division of Biostatistics and Computational BiologyUniversity of Iowa College of DentistryIowa CityIA52242USA
| | - Shaoping Zhang
- Iowa Institute for Oral Health ResearchUniversity of Iowa College of DentistryIowa CityIA52242USA
- Department of PeriodonticsUniversity of Iowa College of DentistryIowa CityIA52242USA
| | - Hongli Sun
- Iowa Institute for Oral Health ResearchUniversity of Iowa College of DentistryIowa CityIA52242USA
- Department of Oral and Maxillofacial SurgeryUniversity of Iowa College of DentistryIowa CityIA52242USA
- Roy J. Carver Department of Biomedical EngineeringUniversity of Iowa College of EngineeringIowa CityIA52242USA
| |
Collapse
|
5
|
Dong ZL, Jiao X, Wang ZG, Yuan K, Yang YQ, Wang Y, Li YT, Wang TC, Kan TY, Wang J, Tao HR. D-mannose alleviates intervertebral disc degeneration through glutamine metabolism. Mil Med Res 2024; 11:28. [PMID: 38711073 PMCID: PMC11071241 DOI: 10.1186/s40779-024-00529-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 04/11/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is a multifaceted condition characterized by heterogeneity, wherein the balance between catabolism and anabolism in the extracellular matrix of nucleus pulposus (NP) cells plays a central role. Presently, the available treatments primarily focus on relieving symptoms associated with IVDD without offering an effective cure targeting its underlying pathophysiological processes. D-mannose (referred to as mannose) has demonstrated anti-catabolic properties in various diseases. Nevertheless, its therapeutic potential in IVDD has yet to be explored. METHODS The study began with optimizing the mannose concentration for restoring NP cells. Transcriptomic analyses were employed to identify the mediators influenced by mannose, with the thioredoxin-interacting protein (Txnip) gene showing the most significant differences. Subsequently, small interfering RNA (siRNA) technology was used to demonstrate that Txnip is the key gene through which mannose exerts its effects. Techniques such as colocalization analysis, molecular docking, and overexpression assays further confirmed the direct regulatory relationship between mannose and TXNIP. To elucidate the mechanism of action of mannose, metabolomics techniques were employed to pinpoint glutamine as a core metabolite affected by mannose. Next, various methods, including integrated omics data and the Gene Expression Omnibus (GEO) database, were used to validate the one-way pathway through which TXNIP regulates glutamine. Finally, the therapeutic effect of mannose on IVDD was validated, elucidating the mechanistic role of TXNIP in glutamine metabolism in both intradiscal and orally treated rats. RESULTS In both in vivo and in vitro experiments, it was discovered that mannose has potent efficacy in alleviating IVDD by inhibiting catabolism. From a mechanistic standpoint, it was shown that mannose exerts its anti-catabolic effects by directly targeting the transcription factor max-like protein X-interacting protein (MondoA), resulting in the upregulation of TXNIP. This upregulation, in turn, inhibits glutamine metabolism, ultimately accomplishing its anti-catabolic effects by suppressing the mitogen-activated protein kinase (MAPK) pathway. More importantly, in vivo experiments have further demonstrated that compared with intradiscal injections, oral administration of mannose at safe concentrations can achieve effective therapeutic outcomes. CONCLUSIONS In summary, through integrated multiomics analysis, including both in vivo and in vitro experiments, this study demonstrated that mannose primarily exerts its anti-catabolic effects on IVDD through the TXNIP-glutamine axis. These findings provide strong evidence supporting the potential of the use of mannose in clinical applications for alleviating IVDD. Compared to existing clinically invasive or pain-relieving therapies for IVDD, the oral administration of mannose has characteristics that are more advantageous for clinical IVDD treatment.
Collapse
Affiliation(s)
- Zheng-Lin Dong
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xin Jiao
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zeng-Guang Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Kai Yuan
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yi-Qi Yang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yao Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yun-Tao Li
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Tian-Chang Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Tian-You Kan
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jian Wang
- School of Medicine, Shanghai University, Shanghai, 200444, China.
| | - Hai-Rong Tao
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
6
|
Bravo Iniguez A, Du M, Zhu MJ. α-Ketoglutarate for Preventing and Managing Intestinal Epithelial Dysfunction. Adv Nutr 2024; 15:100200. [PMID: 38438107 PMCID: PMC11016550 DOI: 10.1016/j.advnut.2024.100200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024] Open
Abstract
The epithelium lining the intestinal tract serves a multifaceted role. It plays a crucial role in nutrient absorption and immune regulation and also acts as a protective barrier, separating underlying tissues from the gut lumen content. Disruptions in the delicate balance of the gut epithelium trigger inflammatory responses, aggravate conditions such as inflammatory bowel disease, and potentially lead to more severe complications such as colorectal cancer. Maintaining intestinal epithelial homeostasis is vital for overall health, and there is growing interest in identifying nutraceuticals that can strengthen the intestinal epithelium. α-Ketoglutarate, a metabolite of the tricarboxylic acid cycle, displays a variety of bioactive effects, including functioning as an antioxidant, a necessary cofactor for epigenetic modification, and exerting anti-inflammatory effects. This article presents a comprehensive overview of studies investigating the potential of α-ketoglutarate supplementation in preventing dysfunction of the intestinal epithelium.
Collapse
Affiliation(s)
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA, United States
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA, United States.
| |
Collapse
|
7
|
Agarwal S, Ghosh R, Verma G, Khadgawat R, Guchhait P. Alpha-ketoglutarate supplementation reduces inflammation and thrombosis in type 2 diabetes by suppressing leukocyte and platelet activation. Clin Exp Immunol 2023; 214:197-208. [PMID: 37498307 PMCID: PMC10714189 DOI: 10.1093/cei/uxad086] [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: 04/27/2023] [Revised: 06/23/2023] [Accepted: 07/26/2023] [Indexed: 07/28/2023] Open
Abstract
The interplay between platelets and leukocytes contributes to the pathogenesis of inflammation, thrombosis, and cardiovascular diseases (CVDs) in type 2 diabetes (T2D). Our recent studies described alpha-ketoglutarate (αKG), a Krebs cycle intermediate metabolite as an inhibitor to platelets and leukocytes activation by suppressing phosphorylated-Akt (pAkt) through augmentation of prolyl hydroxylase-2 (PHD2). Dietary supplementation with a pharmacological concentration of αKG significantly inhibited lung inflammation in mice with either SARS-CoV-2 infection or exposed to hypoxia treatment. We therefore investigated if αKG supplementation could suppress hyperactivation of these blood cells and reduce thromboinflammatory complications in T2D. Our study describes that dietary supplementation with αKG (8 mg/100 g body wt. daily) for 7 days significantly reduced the activation of platelets and leukocytes (neutrophils and monocytes), and accumulation of IL1β, TNFα, and IL6 in peripheral blood of T2D mice. αKG also reduced the infiltration of platelets and leukocytes, and accumulation of inflammatory cytokines in lungs by suppressing pAkt and pP65 signaling. In a cross-sectional investigation, our study also described the elevated platelet-leukocyte aggregates and pro-inflammatory cytokines in circulation of T2D patients. T2D platelets and leukocytes showed an increased aggregation and thrombus formation in vitro. Interestingly, a pre-incubation of T2D blood samples with octyl αKG significantly suppressed the activation of these blood cells and ameliorated aggregate/thrombus formation in vitro. Thus, suggesting a potential therapeutic role of αKG against inflammation, thrombosis, and CVDs in T2D.
Collapse
Affiliation(s)
- Sakshi Agarwal
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Riya Ghosh
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Garima Verma
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Rajesh Khadgawat
- Endocrinology & Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| |
Collapse
|
8
|
Liu H, Ren X, Li Y, Cao Q, Yang L, Jiang S, Fu J, Gao J, Yan L, Li J, Yang W. Effects of different wheat bran fermentation sources on growth performance, nutrient digestibility, serum antioxidant capacity and fecal microbiota in growing pigs. Front Vet Sci 2023; 10:1289010. [PMID: 38033646 PMCID: PMC10687151 DOI: 10.3389/fvets.2023.1289010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023] Open
Abstract
The present study aimed to evaluate the application of different wheat bran fermentation sources in growing pigs. A total of 320 pigs (43 ± 0.21 kg), were randomly allocated to 5 groups in a 21-d trial. The control group was fed a basal diet (CON) containing raw wheat bran, and the other four treatments were fed the diets in which the raw wheat bran in the basal diet was substituted with Aspergillus niger (WBA), Bacillus licheniformis (WBB), Candida utilis (WBC), and Lactobacillus plantarum (WBL) fermented wheat bran, respectively. The results showed that compared to the CON group, the crude fiber and pH values were decreased (p < 0.05), while the gross energy (GE), crude protein (CP), and lactic acid values were increased (p < 0.05) in all the wheat bran fermented by different strains. Compared with other treatments, feeding B. licheniformis fermented wheat bran had higher final weight, average daily gain, as well as lower feed-to-gain ratio. Compared with CON group, pigs fed with fermented wheat bran diets had higher dry matter, CP, and GE availability, serum total protein, albumin and superoxide dismutase levels, and fecal Lactobacillus counts, as well as lower malondialdehyde level and fecal Escherichia coli count. Collectively, our findings suggested that feeding fermented wheat bran, especially B. licheniformis fermented wheat bran, showed beneficial effects on the growth performance, nutrient digestibility, serum antioxidant capacity, and the gut microbiota structure of growing pigs.
Collapse
Affiliation(s)
- Heng Liu
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Xiaojie Ren
- Shandong Taishan Shengliyuan Group Co., Ltd., Tai’an, China
| | - Yang Li
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | | | - Lijie Yang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Shuzhen Jiang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Jiawei Fu
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Jie Gao
- College of Biology and Brewing Engineering, Mount Taishan College, Tai’an, China
| | - Lei Yan
- Shandong New Hope Liuhe Group Co., Ltd., Qingdao, China
| | - Junxun Li
- Shandong Taishan Shengliyuan Group Co., Ltd., Tai’an, China
| | - Weiren Yang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| |
Collapse
|
9
|
Gusdon AM, Fu C, Putluri V, Paz AS, Chen H, Ren X, Hassan MK, Dash P, Coarfa C, Putluri N, Choi HA, Savarraj JPJ. Early Systemic Glycolytic Shift After Aneurysmal Subarachnoid Hemorrhage is Associated with Functional Outcomes. Neurocrit Care 2022; 37:724-734. [PMID: 35799091 PMCID: PMC10473383 DOI: 10.1007/s12028-022-01546-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/03/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Aneurysmal subarachnoid hemorrhage (aSAH) leads to a robust systemic inflammatory response. We hypothesized that an early systemic glycolytic shift occurs after aSAH, resulting in a unique metabolic signature and affecting systemic inflammation. METHODS Control patients and patients with aSAH were analyzed. Samples from patients with aSAH were collected within 24 h of aneurysmal rupture. Mass spectrometry-based metabolomics was performed to assess relative abundance of 16 metabolites involved in the tricarboxylic acid cycle, glycolysis, and pentose phosphate pathway. Principal component analysis was used to segregate control patients from patients with aSAH. Dendrograms were developed to depict correlations between metabolites and cytokines. Analytic models predicting functional outcomes were developed, and receiver operating curves were compared. RESULTS A total of 122 patients with aSAH and 38 control patients were included. Patients with aSAH had higher levels of glycolytic metabolites (3-phosphoglycerate/2-phosphoglycerate, lactate) but lower levels of oxidative metabolites (succinate, malate, fumarate, and oxalate). Patients with higher clinical severity (Hunt-Hess Scale score ≥ 4) had higher levels of glyceraldehyde 3-phosphate and citrate but lower levels of α-ketoglutarate and glutamine. Principal component analysis readily segregated control patients from patients with aSAH. Correlation analysis revealed distinct clusters in control patients that were not observed in patients with aSAH. Higher levels of fumarate were associated with good functional outcomes at discharge (odds ratio [OR] 1.76, 95% confidence interval [CI] 1.15-2.82) in multivariable models, whereas higher levels of citrate were associated with poor functional outcomes at discharge (OR 0.36, 95% CI 0.16-0.73) and at 3 months (OR 0.35, 95% CI 0.14-0.81). No associations were found with delayed cerebral ischemia. Levels of α-ketoglutarate and glutamine correlated with lower levels of interleukin-8, whereas fumarate was associated with lower levels of tumor necrosis factor alpha. CONCLUSIONS Aneurysmal subarachnoid hemorrhage results in a unique pattern of plasma metabolites, indicating a shift toward glycolysis. Higher levels of fumarate and lower levels of citrate were associated with better functional outcomes. These metabolites may represent targets to improve metabolism after aSAH.
Collapse
Affiliation(s)
- Aaron M Gusdon
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA.
- Department of Neurobiology and Anatomy, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA.
| | - Chenlian Fu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Biology, Harvey Mudd College, Claremont, CA, USA
| | - Vasanta Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Atzhiry S Paz
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Hua Chen
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Xuefang Ren
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | | | - Pramod Dash
- Department of Neurobiology and Anatomy, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Huimahn A Choi
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Jude P J Savarraj
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
10
|
Tabibzadeh S. Resolving Geroplasticity to the Balance of Rejuvenins and Geriatrins. Aging Dis 2022; 13:1664-1714. [PMID: 36465174 PMCID: PMC9662275 DOI: 10.14336/ad.2022.0414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/14/2022] [Indexed: 09/29/2024] Open
Abstract
According to the cell centric hypotheses, the deficits that drive aging occur within cells by age dependent progressive damage to organelles, telomeres, biologic signaling pathways, bioinformational molecules, and by exhaustion of stem cells. Here, we amend these hypotheses and propose an eco-centric model for geroplasticity (aging plasticity including aging reversal). According to this model, youth and aging are plastic and require constant maintenance, and, respectively, engage a host of endogenous rejuvenating (rejuvenins) and gero-inducing [geriatrin] factors. Aging in this model is akin to atrophy that occurs as a result of damage or withdrawal of trophic factors. Rejuvenins maintain and geriatrins adversely impact cellular homeostasis, cell fitness, and proliferation, stem cell pools, damage response and repair. Rejuvenins reduce and geriatrins increase the age-related disorders, inflammatory signaling, and senescence and adjust the epigenetic clock. When viewed through this perspective, aging can be successfully reversed by supplementation with rejuvenins and by reducing the levels of geriatrins.
Collapse
Affiliation(s)
- Siamak Tabibzadeh
- Frontiers in Bioscience Research Institute in Aging and Cancer, Irvine, CA 92618, USA
| |
Collapse
|
11
|
Yeshi K, Ruscher R, Loukas A, Wangchuk P. Immunomodulatory and biological properties of helminth-derived small molecules: Potential applications in diagnostics and therapeutics. FRONTIERS IN PARASITOLOGY 2022; 1:984152. [PMID: 39816468 PMCID: PMC11731824 DOI: 10.3389/fpara.2022.984152] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2025]
Abstract
Parasitic helminths secrete and excrete a vast array of molecules known to help skew or suppress the host's immune response, thereby establishing a niche for sustained parasite maintenance. Indeed, the immunomodulatory potency of helminths is attributed mainly to excretory/secretory products (ESPs). The ESPs of helminths and the identified small molecules (SM) are reported to have diverse biological and pharmacological properties. The available literature reports only limited metabolites, and the identity of many metabolites remains unknown due to limitations in the identification protocols and helminth-specific compound libraries. Many metabolites are known to be involved in host-parasite interactions and pathogenicity. For example, fatty acids (e.g., stearic acid) detected in the infective stages of helminths are known to have a role in host interaction through facilitating successful penetration and migration inside the host. Moreover, excreted/secreted SM detected in helminth species are found to possess various biological properties, including anti-inflammatory activities, suggesting their potential in developing immunomodulatory drugs. For example, helminths-derived somatic tissue extracts and whole crude ESPs showed anti-inflammatory properties by inhibiting the secretion of proinflammatory cytokines from human peripheral blood mononuclear cells and suppressing the pathology in chemically-induced experimental mice model of colitis. Unlike bigger molecules like proteins, SM are ideal candidates for drug development since they are small structures, malleable, and lack immunogenicity. Future studies should strive toward identifying unknown SM and isolating the under-explored niche of helminth metabolites using the latest metabolomics technologies and associated software, which hold potential keys for finding new diagnostics and novel therapeutics.
Collapse
Affiliation(s)
- Karma Yeshi
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Cairns, QLD, Australia
| | | | | | | |
Collapse
|
12
|
Wu D, Fan Z, Li J, Zhang Y, Xu Q, Wang L, Wang L. Low Protein Diets Supplemented With Alpha-Ketoglutarate Enhance the Growth Performance, Immune Response, and Intestinal Health in Common Carp ( Cyprinus carpio). Front Immunol 2022; 13:915657. [PMID: 35720284 PMCID: PMC9200961 DOI: 10.3389/fimmu.2022.915657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
To investigate the effects of alpha-ketoglutarate (AKG) supplementation in a low protein (LP) diet on the growth performance, immune response, and intestinal health of common carp (Cyprinus carpio), 600 carp were randomly divided into five dietary groups: a normal protein (NP) diet containing 32% crude protein, an LP diet formulated with 28% crude protein, and LP with AKG at 0.4%, 0.8%, and 1.2% (dry matter). After an 8-week trial period, the results demonstrated that an LP diet led to a decrease in performance, immune response, and intestinal barrier function. Compared with the LP group, the final body weight and weight gain rate in the LP+0.4% AKG group were significantly higher, the feed conversion ratio was significantly decreased with the addition of 0.4% and 0.8% AKG. The supplementation with 0.4% and 0.8% AKG markedly increased the activities of T-SOD and GSH-Px, as well as the expression levels of GPX1a and GPX1b relative to the LP group, whereas the MDA content was significantly decreased in the LP+0.4% AKG group. In addition, the expression levels of tight junctions including claudin-3, claudin-7, ZO-1, and MLCK were significantly up-regulated in the LP+0.4% AKG group, and the relative expression levels of the pro-inflammatory factors IL-1β and IL-6α were significantly lower with the addition of 0.4%, 0.8%, and 1.2% AKG. Moreover, the abundance of Proteobacteria in the LP+0.4% AKG group was lower than that in the LP group, and the abundance of Firmicutes and Fusobacteria was higher at the phylum level. The abundance of Citrobacter in the LP+0.4% AKG group was decreased compared to the LP group, while the abundance of Aeromonas was increased at the genus level. In short, the effects of AKG on the intestinal health of the common carp were systematically and comprehensively evaluated from the perspectives of intestinal physical barrier, chemical barrier, biological barrier, and immune barrier. We found that an LP diet supplemented with 0.4% AKG was beneficial to the growth performance and intestinal health of common carp.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Ze Fan
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Jinnan Li
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Yuanyuan Zhang
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Qiyou Xu
- School of Life Science, Huzhou University, Huzhou, China
| | - Liang Wang
- AHP Application Research Institute, Weifang Addeasy Bio-Technology Co., Ltd, Weifang, China
| | - Liansheng Wang
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| |
Collapse
|
13
|
Yuan Y, Zhu C, Wang Y, Sun J, Feng J, Ma Z, Li P, Peng W, Yin C, Xu G, Xu P, Jiang Y, Jiang Q, Shu G. α-Ketoglutaric acid ameliorates hyperglycemia in diabetes by inhibiting hepatic gluconeogenesis via serpina1e signaling. SCIENCE ADVANCES 2022; 8:eabn2879. [PMID: 35507647 PMCID: PMC9067931 DOI: 10.1126/sciadv.abn2879] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/17/2022] [Indexed: 05/13/2023]
Abstract
Previously, we found that α-ketoglutaric acid (AKG) stimulates muscle hypertrophy and fat loss through 2-oxoglutarate receptor 1 (OXGR1). Here, we demonstrated the beneficial effects of AKG on glucose homeostasis in a diet-induced obesity (DIO) mouse model, which are independent of OXGR1. We also showed that AKG effectively decreased blood glucose and hepatic gluconeogenesis in DIO mice. By using transcriptomic and liver-specific serpina1e deletion mouse model, we further demonstrated that liver serpina1e is required for the inhibitory effects of AKG on hepatic gluconeogenesis. Mechanistically, we supported that extracellular AKG binds with a purinergic receptor, P2RX4, to initiate the solute carrier family 25 member 11 (SLC25A11)-dependent nucleus translocation of intracellular AKG and subsequently induces demethylation of lysine 27 on histone 3 (H3K27) in the seprina1e promoter region to decrease hepatic gluconeogenesis. Collectively, these findings reveal an unexpected mechanism for control of hepatic gluconeogenesis using circulating AKG as a signal molecule.
Collapse
Affiliation(s)
- Yexian Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Canjun Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Yongliang Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jia Sun
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jinlong Feng
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Zewei Ma
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Penglin Li
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Wentong Peng
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Cong Yin
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Guli Xu
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Pingwen Xu
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Qingyan Jiang
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Gang Shu
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| |
Collapse
|
14
|
Dutta M, Lim JJ, Cui JY. Pregnane X Receptor and the Gut-Liver Axis: A Recent Update. Drug Metab Dispos 2022; 50:478-491. [PMID: 34862253 PMCID: PMC11022899 DOI: 10.1124/dmd.121.000415] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 12/02/2021] [Indexed: 02/04/2023] Open
Abstract
It is well-known that the pregnane X receptor (PXR)/Nr1i2 is a critical xenobiotic-sensing nuclear receptor enriched in liver and intestine and is responsible for drug-drug interactions, due to its versatile ligand binding domain (LBD) and target genes involved in xenobiotic biotransformation. PXR can be modulated by various xenobiotics including pharmaceuticals, nutraceuticals, dietary factors, and environmental chemicals. Microbial metabolites such as certain secondary bile acids (BAs) and the tryptophan metabolite indole-3-propionic acid (IPA) are endogenous PXR activators. Gut microbiome is increasingly recognized as an important regulator for host xenobiotic biotransformation and intermediary metabolism. PXR regulates and is regulated by the gut-liver axis. This review summarizes recent research advancements leveraging pharmaco- and toxico-metagenomic approaches that have redefined the previous understanding of PXR. Key topics covered in this review include: (1) genome-wide investigations on novel PXR-target genes, novel PXR-DNA interaction patterns, and novel PXR-targeted intestinal bacteria; (2) key PXR-modulating activators and suppressors of exogenous and endogenous sources; (3) novel bidirectional interactions between PXR and gut microbiome under physiologic, pathophysiological, pharmacological, and toxicological conditions; and (4) modifying factors of PXR-signaling including species and sex differences and time (age, critical windows of exposure, and circadian rhythm). The review also discusses critical knowledge gaps and important future research topics centering around PXR. SIGNIFICANCE STATEMENT: This review summarizes recent research advancements leveraging O'mics approaches that have redefined the previous understanding of the xenobiotic-sensing nuclear receptor pregnane X receptor (PXR). Key topics include: (1) genome-wide investigations on novel PXR-targeted host genes and intestinal bacteria as well as novel PXR-DNA interaction patterns; (2) key PXR modulators including microbial metabolites under physiological, pathophysiological, pharmacological, and toxicological conditions; and (3) modifying factors including species, sex, and time.
Collapse
Affiliation(s)
- Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
15
|
Mimicking caloric restriction for anti-aging effects: the pro-oxidant role of alpha-ketoglutarate. CURRENT OPINION IN TOXICOLOGY 2022. [DOI: 10.1016/j.cotox.2022.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
16
|
Wang L, Fan Z, Zhang Y, Wu D, Li J, Xu Q. Effect of phosphorus on growth performance, intestinal tight junctions, Nrf2 signaling pathway and immune response of juvenile mirror carp (Cyprinus carpio) fed different α-ketoglutarate levels. FISH & SHELLFISH IMMUNOLOGY 2022; 120:271-279. [PMID: 34863945 DOI: 10.1016/j.fsi.2021.11.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 06/13/2023]
Abstract
Previous research has shown that dietary α-ketoglutarate (AKG) supplementation can promote growth performance, phosphorus metabolism, and skeletal development of juvenile mirror carp (Cyprinus carpio) fed low phosphorous diets. In the current study, we tested the hypothesis that 1% AKG dietary supplementation reduces the dietary phosphorus requirements of juvenile mirror carp. A total of 12 experimental isoproteic and isolipidic diets containing available phosphorus levels of 0.21%, 0.38%, 0.55%, 0.72%, 0.89%, and 1.07% dry matter with either 0 or 1% AKG supplementation were used in the study. A total of 1080 juvenile fish of similar initial weight (0.90 ± 0.03 g) were selected and randomly assigned to 36 tanks. There were three replicates for each experimental group, with a density of 30 fish per tank. Fish were fed to satiation for 8 weeks. The results indicated that fish fed the diet supplemented with 1% AKG showed a significant increase in final body weight (FBW), weight gain rate (WGR), feed intake (FI) and phosphorus intake (PI) compared to the diet without AKG (P < 0.05). FBW and WGR increased significantly with increasing available phosphorus levels from 0.21% to 0.89% (P < 0.05). The mRNA expression of ZO-1, claudin 11, and occludin was significantly increased by dietary AKG and phosphorus (P < 0.05). The mRNA expression of Nrf2, GPx1a, and CAT in the Nrf2 signaling pathway was significantly increased by dietary AKG and phosphorus (P < 0.05). The expression levels of IL-10 and TGF-β2 were significantly increased by dietary AKG and phosphorus, but the expression levels of IL-1β, IL-8, IL-10, TNF-a and NF-κB were significantly decreased with dietary AKG and phosphorus supplementation (P < 0.05). Based on second-order polynomial regression analysis of WGR against dietary phosphorus levels, the optimal dietary phosphorus level was found to be 0.79% of dry feed for juvenile mirror carp fed a diet with 1% AKG supplementation and 0.93% of dry feed without AKG supplementation. This study confirmed that AKG supplementation can reduce the phosphorus requirements of juvenile mirror carp by promoting growth performance, intestinal tight junctions, Nrf2 signaling pathways and immune response.
Collapse
Affiliation(s)
- Liansheng Wang
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, 150070, PR China.
| | - Ze Fan
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, 150070, PR China
| | - Yuanyuan Zhang
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, 150070, PR China
| | - Di Wu
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, 150070, PR China
| | - Jinnan Li
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, 150070, PR China
| | - Qiyou Xu
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, 150070, PR China; School of Life Science, Huzhou University, Huzhou, 313000, PR China
| |
Collapse
|
17
|
Wangchuk P, Anderson D, Yeshi K, Loukas A. Identification of Small Molecules of the Infective Stage of Human Hookworm Using LCMS-Based Metabolomics and Lipidomics Protocols. ACS Infect Dis 2021; 7:3264-3276. [PMID: 34767348 DOI: 10.1021/acsinfecdis.1c00428] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hookworm infections affect millions of people worldwide and are responsible for impaired mental and physical growth in children, and anemias. There is no vaccine, and increasing anthelmintic drug resistance in nematodes of domestic animals, and reduced drug cure rates in nematode infections of humans is alarming. Despite this looming health problem, there is a significant knowledge gap in terms of nonproteinaceous "excretory/secretory products" (ESPs) and how they orchestrate a parasitic existence. In the current study, we have conducted the first metabolomic and lipidomic analysis of the infective third-stage filariform larvae (L3) of the predominant human hookworm Necator americanus using liquid chromatography-mass spectrometry. Altogether, we have identified a total of 645 small molecules that were mainly produced through amino acid and glycerophospholipid metabolism. Putatively, 495 metabolites were unique to the somatic tissue extract, and 34 metabolites were present only in the ESP component. More than 21 novel mass features with nitrogen and sulfur functional groups were detected in the ESP component for the first time from helminths. While this study could not establish the biological functions of the metabolites identified, literature searches revealed that these metabolites possess various biological properties, including anti-inflammatory activities. These metabolites are likely used by the parasite upon exposure to a host to facilitate skin penetration, passage through different tissues, and immune regulation in the small bowel. Overall, the results presented herein offer significant insight into the metabolome of N. americanus L3 and have the potential to instigate future work to establish biomarkers of infection. This area urgently needs attention, given the lack of sensitive point-of-care diagnostic tools.
Collapse
Affiliation(s)
- Phurpa Wangchuk
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Road, Smithfield, Cairns, Queensland 4878, Australia
| | - Dovile Anderson
- Monash Institute of Pharmaceutical Sciences, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
| | - Karma Yeshi
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Road, Smithfield, Cairns, Queensland 4878, Australia
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Road, Smithfield, Cairns, Queensland 4878, Australia
| |
Collapse
|
18
|
Tang X, Liu X, Zhang K. Effects of Microbial Fermented Feed on Serum Biochemical Profile, Carcass Traits, Meat Amino Acid and Fatty Acid Profile, and Gut Microbiome Composition of Finishing Pigs. Front Vet Sci 2021; 8:744630. [PMID: 34805337 PMCID: PMC8600044 DOI: 10.3389/fvets.2021.744630] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022] Open
Abstract
Microbial fermented feed is an important part of feed industry, while little research has focused on the solid-state fermentation of complete feed. Herein, the purpose of the present study was to investigate the effects of fermented complete feed (FCF) on the growth performance, biochemical profile, carcass traits, meat proximate composition, meat amino acid and fatty acid profile, and gut microbiome composition of finishing pigs, thereby providing references for the application of FCF in animal production. Twenty Duroc × Landrace × Yorkshire pigs with an average body weight (BW) of 48.74 ± 1.49 kg were divided randomly into control group (pigs received a basal diet, CN, n = 10) and FCF group (pigs fed with FCF, n = 10). The experiment lasted for 60 days. FCF improved the growth performance, which was indicated by a significantly increased final BW, average daily gain and average daily feed intake, and a significantly decreased feed-to-gain ratio. FCF improved biochemical profile, which was indicated by a higher alkaline phosphatase, glucose, immunoglobulin G, immunoglobulin M, superoxide dismutase, and total antioxidant capacity content. Pigs that received FCF had better carcass traits and meat quality than did pigs that received basal diet, which was indicated by a higher carcass length, crude protein content, lysine content, Glu content, C18:ln9c, C18:2n6c, C20:4n6, and unsaturated fatty acid content and a lower average back-fat thickness, C18:0, and saturated fatty acids. FCF significantly reduced the relative abundances of presumably pathogenic bacteria of phylum Proteobacteria and genus Escherichia–Shigella and enhanced the relative abundances of likely beneficial bacteria of phylum Firmicutes and genus Clostridium. In summary, FCF had a certain effect on the improvement of growth performance, serum biochemical profile, carcass traits, meat proximate composition, amino acid and fatty acid profile, and gut microbiome composition of finishing pigs.
Collapse
Affiliation(s)
- Xiaopeng Tang
- State Engineering Technology Institute for Karst Desertfication Control, School of Karst Science, Guizhou Normal University, Guiyang, China
| | - Xuguang Liu
- State Engineering Technology Institute for Karst Desertfication Control, School of Karst Science, Guizhou Normal University, Guiyang, China
| | - Kai Zhang
- College of Animal Science, Shanxi Agricultural University, Jinzhong, China
| |
Collapse
|
19
|
Shrimali NM, Agarwal S, Kaur S, Bhattacharya S, Bhattacharyya S, Prchal JT, Guchhait P. α-Ketoglutarate Inhibits Thrombosis and Inflammation by Prolyl Hydroxylase-2 Mediated Inactivation of Phospho-Akt. EBioMedicine 2021; 73:103672. [PMID: 34740102 PMCID: PMC8579134 DOI: 10.1016/j.ebiom.2021.103672] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 12/27/2022] Open
Abstract
Background Phospho-Akt1 (pAkt1) undergoes prolyl hydroxylation at Pro125 and Pro313 by the prolyl hydroxylase-2 (PHD2) in a reaction decarboxylating α-ketoglutarate (αKG). We investigated whether the αKG supplementation could inhibit Akt-mediated activation of platelets and monocytes, in vitro as well as in vivo, by augmenting PHD2 activity. Methods We treated platelets or monocytes isolated from healthy individuals with αKG in presence of agonists in vitro and assessed the signalling molecules including pAkt1. We supplemented mice with dietary αKG and estimated the functional responses of platelets and monocytes ex vivo. Further, we investigated the impact of dietary αKG on inflammation and thrombosis in lungs of mice either treated with thrombosis-inducing agent carrageenan or infected with SARS-CoV-2. Findings Octyl αKG supplementation to platelets promoted PHD2 activity through elevated intracellular αKG to succinate ratio, and reduced aggregation in vitro by suppressing pAkt1(Thr308). Augmented PHD2 activity was confirmed by increased hydroxylated-proline and enhanced binding of PHD2 to pAkt in αKG-treated platelets. Contrastingly, inhibitors of PHD2 significantly increased pAkt1 in platelets. Octyl-αKG followed similar mechanism in monocytes to inhibit cytokine secretion in vitro. Our data also describe a suppressed pAkt1 and reduced activation of platelets and leukocytes ex vivo from mice supplemented with dietary αKG, unaccompanied by alteration in their number. Dietary αKG significantly reduced clot formation and leukocyte accumulation in various organs including lungs of mice treated with thrombosis-inducing agent carrageenan. Importantly, in SARS-CoV-2 infected hamsters, we observed a significant rescue effect of dietary αKG on inflamed lungs with significantly reduced leukocyte accumulation, clot formation and viral load alongside down-modulation of pAkt in the lung of the infected animals. Interpretation Our study suggests that dietary αKG supplementation prevents Akt-driven maladies such as thrombosis and inflammation and rescues pathology of COVID19-infected lungs. Funding Study was funded by the Department of Biotechnology (DBT), Govt. of India (grants: BT/PR22881 and BT/PR22985); and the Science and Engineering Research Board, Govt. of India (CRG/000092).
Collapse
Affiliation(s)
- Nishith M Shrimali
- Regional Centre for Biotechnology; National Capital Region Biotech Science Cluster, Faridabad, India
| | - Sakshi Agarwal
- Regional Centre for Biotechnology; National Capital Region Biotech Science Cluster, Faridabad, India
| | - Simrandeep Kaur
- Regional Centre for Biotechnology; National Capital Region Biotech Science Cluster, Faridabad, India
| | - Sulagna Bhattacharya
- Regional Centre for Biotechnology; National Capital Region Biotech Science Cluster, Faridabad, India
| | - Sankar Bhattacharyya
- Translational Health Science Technology Institute; National Capital Region Biotech Science Cluster, Faridabad, India
| | - Josef T Prchal
- Department of Medicine, University of Utah School of Medicine & Huntsman Cancer Center and George E. Whalen Veteran's Administration Medical Center, Salt Lake City, UT, USA
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology; National Capital Region Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
20
|
Wu D, Fan Z, Li J, Zhang Y, Wang C, Xu Q, Wang L. Evaluation of Alpha-Ketoglutarate Supplementation on the Improvement of Intestinal Antioxidant Capacity and Immune Response in Songpu Mirror Carp ( Cyprinus carpio) After Infection With Aeromonas hydrophila. Front Immunol 2021; 12:690234. [PMID: 34220849 PMCID: PMC8250152 DOI: 10.3389/fimmu.2021.690234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/04/2021] [Indexed: 11/24/2022] Open
Abstract
As an intermediate substance of the tricarboxylic acid cycle and a precursor substance of glutamic acid synthesis, the effect of alpha-ketoglutarate on growth and protein synthesis has been extensively studied. However, its prevention and treatment of pathogenic bacteria and its mechanism have not yet been noticed. To evaluate the effects of alpha-ketoglutarate on intestinal antioxidant capacity and immune response of Songpu mirror carp, a total of 360 fish with an average initial weight of 6.54 ± 0.08 g were fed diets containing alpha-ketoglutarate with 1% for 8 weeks. At the end of the feeding trial, the fish were challenged with Aeromonas hydrophila for 2 weeks. The results indicated that alpha-ketoglutarate supplementation significantly increased the survival rate of carp after infection with Aeromonas hydrophila (P < 0.05), and the contents of immune digestion enzymes including lysozyme, alkaline phosphatase and the concentration of complement C4 were markedly enhanced after alpha-ketoglutarate supplementation (P < 0.05). Also, appropriate alpha-ketoglutarate increased the activities of total antioxidant capacity and catalase and prevented the up-regulation in the mRNA expression levels of pro-inflammatory cytokines including tumor necrosis factor-α, interleukin-1β, interleukin-6, and interleukin-8 (P < 0.05). Furthermore, the mRNA expression levels of toll-like receptor 4 (TLR4), and nuclear factor kappa-B (NF-κB) were strikingly increased after infection with Aeromonas hydrophila (P < 0.05), while the TLR4 was strikingly decreased with alpha-ketoglutarate supplementation (P < 0.05). Moreover, the mRNA expression levels of tight junctions including claudin-1, claudin-3, claudin-7, claudin-11 and myosin light chain kinases (MLCK) were upregulated after alpha-ketoglutarate supplementation (P < 0.05). In summary, the appropriate alpha-ketoglutarate supplementation could increase survival rate, strengthen the intestinal enzyme immunosuppressive activities, antioxidant capacities and alleviate the intestinal inflammation, thereby promoting the intestinal immune responses and barrier functions of Songpu mirror carp via activating TLR4/MyD88/NF-κB and MLCK signaling pathways after infection with Aeromonas hydrophila.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Ze Fan
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Jinnan Li
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Yuanyuan Zhang
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Chang'an Wang
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Qiyou Xu
- School of Life Science, Huzhou University, Huzhou, China
| | - Liansheng Wang
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| |
Collapse
|
21
|
Peng P, Deng D, Chen S, Li C, Luo J, Romeo A, Li T, Tang X, Fang R. The Effects of Dietary Porous Zinc Oxide Supplementation on Growth Performance, Inflammatory Cytokines and Tight Junction's Gene Expression in Early-Weaned Piglets. J Nutr Sci Vitaminol (Tokyo) 2021; 66:311-318. [PMID: 32863303 DOI: 10.3177/jnsv.66.311] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
This study was conducted to investigate the effect of dietary porous ZnO supplementation on the growth performance, inflammatory cytokines and tight junction's gene expression in weaned piglets. A total of 192 weaned piglets were randomly allocated to 4 experimental groups (n=48/group) and fed, during 14 d, with one of the following dietary treatments: 1) basal diet (NC); 2) basal diet with 3,000 mg/kg of conventional ZnO (PC); 3) basal diet with 750 mg/kg of porous ZnO (low inclusion porous ZnO, LP-ZnO); 4) basal diet with 1,500 mg/kg porous ZnO (high inclusion porous ZnO, HP-ZnO). Results showed that dietary supplementation with regular ZnO or porous ZnO (750 and 1,500 mg/kg) improved average daily gain (ADG), feed to gain ratio (F/G) and jejunum morphology, while decreasing diarrhea incidence. Compared with the NC group, porous ZnO at both doses (750 or 1,500 mg/kg) increased serum alkaline phosphatase (ALP), immunoglobulin G (IgG) and insulin-like growth factor 1 (IGF-1) concentrations, but decreased serum glucose (GLU). Moreover, the mRNA expression of anti-inflammation cytokine (TGF-β), tight junction (Occludin, ZO-1) in the jejunum by different ZnO administration were significantly increased compared with the NC group, while mRNA expression of pro-inflammatory (IL-8), membrane channels that transport water (AQP3) and miR-122a were significantly decreased. It can be concluded that porous ZnO even at low dose (750 mg/kg) can be an effective alternative to pharmacological (3,000 mg/kg) conventional ZnO in reducing diarrhea, promoting the growth performance, increasing anti-inflammatory cytokines and tight junctions, reducing pro-inflammatory cytokines of weaned piglets.
Collapse
Affiliation(s)
- Peng Peng
- College of Animal Science and Technology, Hunan Agriculture University.,Tangrenshen Group, Liyu Industry Park, National High-tech Development Area
| | - Dun Deng
- Tangrenshen Group, Liyu Industry Park, National High-tech Development Area
| | - Sijia Chen
- College of Animal Science and Technology, Hunan Agriculture University
| | - Chengliang Li
- College of Animal Science and Technology, Hunan Agriculture University
| | - Jie Luo
- Tangrenshen Group, Liyu Industry Park, National High-tech Development Area
| | | | - Tiejun Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production
| | - Xiaopeng Tang
- State Key Laboratory Cultivation for Karst Mountain Ecology Environment of Guizhou Province, School of Karst Science, Guizhou Normal University
| | - Rejun Fang
- College of Animal Science and Technology, Hunan Agriculture University
| |
Collapse
|
22
|
Tian Q, Bravo Iniguez A, Sun Q, Wang H, Du M, Zhu MJ. Dietary Alpha-Ketoglutarate Promotes Epithelial Metabolic Transition and Protects against DSS-Induced Colitis. Mol Nutr Food Res 2021; 65:e2000936. [PMID: 33547710 DOI: 10.1002/mnfr.202000936] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/12/2020] [Indexed: 12/13/2022]
Abstract
SCOPE As a natural compound in foods, alpha-ketoglutarate (aKG) is one of the key metabolites maintaining energy homeostasis. This study examines the beneficial effects of dietary aKG against the development of experimental colitis and further explores the underlying molecular mechanisms. METHODS AND RESULTS Eight-week-old male C57BL/6 mice receive drinking water with or without 1% aKG for 4 weeks. At week 3, colitis is induced by 2.5% dextran sulfate sodium (DSS) for 7 days followed by 7 days recovery. Dietary aKG supplementation decreases DSS-induced body weight loss, gross bleeding, fecal consistency score, and disease activity index. In agreement, aKG supplementation restores DSS-associated colon shortening, ameliorated mucosal damage, and macrophage infiltration into colonic tissue, which are associated with suppressed gut inflammation and Wnt signaling, and improved epithelial structure. Consistently, aKG supplementation enhances M1 to M2 macrophage polarization and strengthens intestinal barrier function. Additionally, aKG supplementation elevates colonic aKG levels while decreasing 2-hydroxyglutarate levels, which increases oxidative instead of glycolytic metabolism. CONCLUSION aKG supplementation protects against epithelial damage and ameliorates DSS-induced colitis, which are associated with suppressed inflammation, Wnt signaling pathway, and glycolysis. Intake of foods enriched with aKG or aKG supplementation can be an alternative approach for the prevention or treatment of colitis that are common in Western societies.
Collapse
Affiliation(s)
- Qiyu Tian
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
- Department of Animal Science, Washington State University, Pullman, WA, 99164, USA
| | | | - Qi Sun
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
| | - Hongbin Wang
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
| | - Min Du
- Department of Animal Science, Washington State University, Pullman, WA, 99164, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
| |
Collapse
|
23
|
Li Y, Li X, Gao Y, Huang C, Lin D. NMR-Based Metabolomic Analysis for the Effects of α-Ketoglutarate Supplementation on C2C12 Myoblasts in Different Energy States. Molecules 2021; 26:1841. [PMID: 33805924 PMCID: PMC8037044 DOI: 10.3390/molecules26071841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 11/17/2022] Open
Abstract
α-Ketoglutarate (AKG) is attracting much attention from researchers owing to its beneficial effects on anti-aging and cancer suppression, and, more recently, in nutritional supplements. Given that glucose is the main source of energy to maintain normal physiological functions of skeletal muscle, the effects of AKG supplementation for improving muscle performance are closely related to the glucose level in skeletal muscle. The differences of AKG-induced effects in skeletal muscle between two states of normal energy and energy deficiency are unclear. Furthermore, AKG-induced metabolic changes in skeletal muscles in different energy states also remain elusive. Here, we assessed the effects of AKG supplementation on mouse C2C12 myoblast cells cultured both in normal medium (Nor cells) and in low-glucose medium (Low cells), which were used to mimic two states of normal energy and energy deficiency, respectively. We further performed NMR-based metabolomic analysis to address AKG-induced metabolic changes in Nor and Low cells. AKG supplementation significantly promoted the proliferation and differentiation of cells in the two energy states through glutamine metabolism, oxidative stress, and energy metabolism. Under normal culture conditions, AKG up-regulated the intracellular glutamine level, changed the cellular energy status, and maintained the antioxidant capacity of cells. Under low-glucose culture condition, AKG served as a metabolic substrate to reduce the glutamine-dependence of cells, remarkably enhanced the antioxidant capacity of cells and significantly elevated the intracellular ATP level, thereby ensuring the normal growth and metabolism of cells in the state of energy deficiency. Our results provide a mechanistic understanding of the effects of AKG supplements on myoblasts in both normal energy and energy deficiency states. This work may be beneficial to the exploitation of AKG applications in clinical treatments and nutritional supplementations.
Collapse
Affiliation(s)
- Yantong Li
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| | - Xiaoyuan Li
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| | - Yifeng Gao
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361021, China
| | - Donghai Lin
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| |
Collapse
|
24
|
Pleiotropic effects of alpha-ketoglutarate as a potential anti-ageing agent. Ageing Res Rev 2021; 66:101237. [PMID: 33340716 DOI: 10.1016/j.arr.2020.101237] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/23/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
An intermediate of tricarboxylic acid cycle alpha-ketoglutarate (AKG) is involved in pleiotropic metabolic and regulatory pathways in the cell, including energy production, biosynthesis of certain amino acids, collagen biosynthesis, epigenetic regulation of gene expression, regulation of redox homeostasis, and detoxification of hazardous substances. Recently, AKG supplement was found to extend lifespan and delay the onset of age-associated decline in experimental models such as nematodes, fruit flies, yeasts, and mice. This review summarizes current knowledge on metabolic and regulatory functions of AKG and its potential anti-ageing effects. Impact on epigenetic regulation of ageing via being an obligate substrate of DNA and histone demethylases, direct antioxidant properties, and function as mimetic of caloric restriction and hormesis-induced agent are among proposed mechanisms of AKG geroprotective action. Due to influence on mitochondrial respiration, AKG can stimulate production of reactive oxygen species (ROS) by mitochondria. According to hormesis hypothesis, moderate stimulation of ROS production could have rather beneficial biological effects, than detrimental ones, because of the induction of defensive mechanisms that improve resistance to stressors and age-related diseases and slow down functional senescence. Discrepancies found in different models and limitations of AKG as a geroprotective drug are discussed.
Collapse
|
25
|
Fu C, Yang Y, Kumrungsee T, Kimoto A, Izu H, Kato N. Low-Dose Ethanol Has Impacts on Plasma Levels of Metabolites Relating to Chronic Disease Risk in SAMP8 mice. J Nutr Sci Vitaminol (Tokyo) 2021; 66:553-560. [PMID: 33390397 DOI: 10.3177/jnsv.66.553] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The effects of low-dose alcohol on experimental animals are unclear. This study examined plasma metabolites in senescence-accelerated mice 8 (SAMP8) given low-dose ethanol, and compared them with aging progress and skeletal muscle strength. Male SAMP8 mice (10-wk-old) were given drinking water containing 0% (control), 1%, 2%, or 5% (v/v) ethanol for 14 wk. Compared with the control group, only mice who consumed 1% ethanol experienced a lower senescence score at 18 and 23 wk, as well as an increased limb grip strength at 21 wk. Plasma metabolites of control, 1% and 2% ethanol groups were analyzed by capillary electrophoresis-time-of-flight mass spectrometry (CE-TOF/MS). Among the 7 metabolites affected by ethanol, notewhorthy is the positive association of the ethanol levels in drinking water with the levels of α-ketoglutarate (antioxidant and anti-inflammatory metabolite) and hippurate (antioxidant and microbial co-metabolite) (p<0.05). Intriguingly, the levels of 2-hydroxyisobutyrate (the biomarker of energy metabolism and microbial co-metabolite) were higher in the 1% ethanol group (p<0.05), but not in the 2% ethanol group as compared to the control. Furthermore, the levels of some of the metabolites affected were correlated with some variables in the grading score of senescence and muscle strength. This study provides a novel insight into how low-dose ethanol in SAMP8 mice modulates the levels of circulating metabolites relating to chronic disease risk.
Collapse
Affiliation(s)
- Churan Fu
- Graduate School of Integrated Sciences for Life, Hiroshima University
| | - Yongshou Yang
- Graduate School of Integrated Sciences for Life, Hiroshima University
| | | | - Akiko Kimoto
- Faculty of Human Ecology, Yasuda Women's University
| | - Hanae Izu
- Quality and Evaluation Research Division, National Research Institute of Brewing
| | - Norihisa Kato
- Graduate School of Integrated Sciences for Life, Hiroshima University
| |
Collapse
|
26
|
Yeshi K, Creek DJ, Anderson D, Ritmejerytė E, Becker L, Loukas A, Wangchuk P. Metabolomes and Lipidomes of the Infective Stages of the Gastrointestinal nematodes, Nippostrongylus brasiliensis and Trichuris muris. Metabolites 2020; 10:metabo10110446. [PMID: 33171998 PMCID: PMC7694664 DOI: 10.3390/metabo10110446] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 02/08/2023] Open
Abstract
Soil-transmitted helminths, including hookworms and whipworms, infect billions of people worldwide. Their capacity to penetrate and migrate through their hosts’ tissues is influenced by the suite of molecules produced by the infective developmental stages. To facilitate a better understanding of the immunobiology and pathogenicity of human hookworms and whipworms, we investigated the metabolomes of the infective stage of Nippostrongylus brasiliensis third-stage larvae (L3) which penetrate the skin and Trichuris muris eggs which are orally ingested, using untargeted liquid chromatography-mass spectrometry (LC-MS). We identified 55 polar metabolites through Metabolomics Standard Initiative level-1 (MSI-I) identification from N. brasiliensis and T. muris infective stages, out of which seven were unique to excretory/secretory products (ESPs) of N. brasiliensis L3. Amino acids were a principal constituent (33 amino acids). Additionally, we identified 350 putative lipids, out of which 28 (all known lipids) were unique to N. brasiliensis L3 somatic extract and four to T. muris embryonated egg somatic extract. Glycerophospholipids and glycerolipids were the major lipid groups. The catalogue of metabolites identified in this study shed light on the biology, and possible therapeutic and diagnostic targets for the treatment of these critical infectious pathogens. Moreover, with the growing body of literature on the therapeutic utility of helminth ESPs for treating inflammatory diseases, a role for metabolites is likely but has received little attention thus far.
Collapse
Affiliation(s)
- Karma Yeshi
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; (E.R.); (L.B.); (A.L.)
- Correspondence: (K.Y.); (P.W.)
| | - Darren J. Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia; (D.J.C.); (D.A.)
| | - Dovile Anderson
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia; (D.J.C.); (D.A.)
| | - Edita Ritmejerytė
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; (E.R.); (L.B.); (A.L.)
| | - Luke Becker
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; (E.R.); (L.B.); (A.L.)
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; (E.R.); (L.B.); (A.L.)
| | - Phurpa Wangchuk
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; (E.R.); (L.B.); (A.L.)
- Correspondence: (K.Y.); (P.W.)
| |
Collapse
|
27
|
Daujat-Chavanieu M, Gerbal-Chaloin S. Regulation of CAR and PXR Expression in Health and Disease. Cells 2020; 9:E2395. [PMID: 33142929 PMCID: PMC7692647 DOI: 10.3390/cells9112395] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/22/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Pregnane X receptor (PXR, NR1I2) and constitutive androstane receptor (CAR, NR1I3) are members of the nuclear receptor superfamily that mainly act as ligand-activated transcription factors. Their functions have long been associated with the regulation of drug metabolism and disposition, and it is now well established that they are implicated in physiological and pathological conditions. Considerable efforts have been made to understand the regulation of their activity by their cognate ligand; however, additional regulatory mechanisms, among which the regulation of their expression, modulate their pleiotropic effects. This review summarizes the current knowledge on CAR and PXR expression during development and adult life; tissue distribution; spatial, temporal, and metabolic regulations; as well as in pathological situations, including chronic diseases and cancers. The expression of CAR and PXR is modulated by complex regulatory mechanisms that involve the interplay of transcription factors and also post-transcriptional and epigenetic modifications. Moreover, many environmental stimuli affect CAR and PXR expression through mechanisms that have not been elucidated.
Collapse
Affiliation(s)
| | - Sabine Gerbal-Chaloin
- IRMB, University of Montpellier, INSERM, CHU Montpellier, 34295 Montpellier, France;
| |
Collapse
|
28
|
Rasmussen MK. Porcine cytochrome P450 3A: current status on expression and regulation. Arch Toxicol 2020; 94:1899-1914. [PMID: 32172306 DOI: 10.1007/s00204-020-02710-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Abstract
The cytochrome P450s (CYPs) constitute a family of enzymes maintaining vital functions in the body and are mostly recognized for their significant role in detoxification. Of the CYP subfamilies, CYP3A, is one of the most active in the clearance of drugs and other xenobiotics. During the last decades, much focus has been on exploring different models for human CYP3A regulation, expression and activity. In that respect, the growing knowledge of the porcine CYP3As is of great interest. Although many aspects of porcine CYP3A regulation and activity are still unknown, the current literature provides a basic understanding of the porcine CYP3As that can be used e.g., when translating results from studies done in the porcine model into human settings. In this review, the current knowledge about porcine CYP3A expression, regulation, activity and metabolic significance are highlighted. Future research needs are also identified.
Collapse
|
29
|
Zhang J, Zhao Z, Bai H, Wang M, Jiao L, Peng W, Wu T, Liu T, Chen H, Song X, Wu L, Hu X, Wu Q, Zhou J, Song J, Lyv M, Ying B. Genetic polymorphisms in PXR and NF-κB1 influence susceptibility to anti-tuberculosis drug-induced liver injury. PLoS One 2019; 14:e0222033. [PMID: 31490979 PMCID: PMC6730870 DOI: 10.1371/journal.pone.0222033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/20/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Pregnane X receptor (PXR) regulates the expression of drug-metabolizing enzymes and transport enzymes. NF-κB not only plays a role in liver homeostasis and injury-healing processes by regulating inflammatory responses but may also regulate the transcription of PXR. Currently, genetic polymorphisms in PXR are associated with adverse drug effects. Because little is known about the association between NF-κB1 genetic polymorphisms and adverse drug reactions, we explored the association between PXR and NF-κB1 single nucleotide polymorphisms (SNPs) and susceptibility to anti-tuberculosis drug-induced liver injury (ATDILI). MATERIALS AND METHODS A total of 746 tuberculosis patients (118 with ATDILI and 628 without ATDILI) were prospectively enrolled at West China Hospital between December 2014 and April 2018. Nine selected SNPs (rs3814055, rs13059232, rs7643645 and rs3732360 in PXR and rs78872571, rs4647992, rs60371688, rs1598861 and rs3774959 in NF-κB1) were genotyped with a custom-designed 2x48-plex SNP Scan TM Kit. The frequencies of the alleles, genotypes and genetic models of the variants were compared between patients with or without ATDILI, while joint effect analysis of the SNP-SNP interactions was performed using multiplicative and additive models. The odds ratios (ORs) and the corresponding 95% confidence intervals (CIs) were calculated. RESULTS The T allele of rs3814055 in PXR was associated with a decreased risk for ATDILI (OR 0.61; 95% CI: 0.42-0.89, p = 0.0098). The T alleles of rs78872571 and rs4647992 in NF-κB1 were significantly associated with an increased risk for ATDILI (OR 1.91; 95% CI: 1.06-3.43, p = 0.028 and OR 1.81; 1.06-3.10, p = 0.029, respectively). The allele, genotype and genetic model frequencies were similar in the two groups for the other six SNPs (all P>0.05). There were no multiplicative or additive interactions between the SNPs. CONCLUSION Our study is the first to reveal that rs3814055 variants in PXR and rs78872571 and rs4647992 variants in NF-κB1 are associated with susceptibility to ATDILI caused by first-line anti-tuberculosis combination treatment in the Han Chinese population.
Collapse
Affiliation(s)
- Jingwei Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Zhenzhen Zhao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Hao Bai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Minjin Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Lin Jiao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Wu Peng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Tao Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Tangyuheng Liu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Hao Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xingbo Song
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Lijuan Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xuejiao Hu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Qian Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Jiajia Song
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Mengyuan Lyv
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- * E-mail:
| |
Collapse
|
30
|
Li S, Fu C, Zhao Y, He J. Intervention with α-Ketoglutarate Ameliorates Colitis-Related Colorectal Carcinoma via Modulation of the Gut Microbiome. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8020785. [PMID: 31317039 PMCID: PMC6601488 DOI: 10.1155/2019/8020785] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 05/18/2019] [Indexed: 12/21/2022]
Abstract
The intestinal microbiome plays a crucial role in promoting intestinal health, and perturbations to its constitution may result in chronic intestinal inflammation and lead to colorectal cancer (CRC). α-Ketoglutarate is an important intermediary in the NF-κB-mediated inflammatory pathway that maintains intestinal homeostasis and prevents initiation of intestinal inflammation, a known precursor to carcinoma development. The objective of this study was to assess the potential protective effects of α-ketoglutarate intervention against CRC development, which may arise due to its known anti-inflammatory and antitumour effects. CRC was induced in C57BL/6 mice using azoxymethane (AOM) and dextran sulfate sodium (DSS). Tumour frequency, histological rating, and colonic microbiota were assessed in colonic samples. The findings demonstrated that α-ketoglutarate offered significant protection against CRC development in mice. Furthermore, α-ketoglutarate also exhibited immunomodulatory effects mediated via downregulation of interleukin (IL)-6, IL-22, tumour necrosis factor (TNF)-α, and IL-1β cytokines. Finally, intervention with α-ketoglutarate tended to minimise the frequency of opportunistic pathogens (Escherichia and Enterococcus) while increasing the populations of Akkermansia, Butyricicoccus, Clostridium, and Ruminococcus. Taken together, our findings show that dietary α-ketoglutarate intervention may protect against inflammation-related CRC.
Collapse
Affiliation(s)
- Si Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Chenxing Fu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Yurong Zhao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Jianhua He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| |
Collapse
|
31
|
He L, Wu J, Tang W, Zhou X, Lin Q, Luo F, Yin Y, Li T. Prevention of Oxidative Stress by α-Ketoglutarate via Activation of CAR Signaling and Modulation of the Expression of Key Antioxidant-Associated Targets in Vivo and in Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:11273-11283. [PMID: 30346763 DOI: 10.1021/acs.jafc.8b04470] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
α-Ketoglutarate (AKG) can act as an antioxidant both in vitro and in vivo. However, the mechanisms of the protective effects of AKG are still not well understood. We evaluated the effects of AKG supplementation on the regulation of the constitutive-androstane-receptor (CAR) pathway in porcine intestinal cells and piglets exposed to H2O2. Our data showed that AKG treatment significantly increased not only the intra- and extracellular levels of AKG (26.9 ± 1.31 μmol/g protein, 1064.4 ± 39.80 μmol/L medium) but also those of Asp (29.3 ± 0.21 μmol/g, 4.20 ± 0.11 μmol/L), Gln (24.82 ± 1.50 μmol/g, 1087.80 ± 16.10 μmol/L), and Glu (91.90 ± 3.6 μmol/g, 19.76 ± 1.00 μmol/L). There was approximately a 4-fold increase in α-ketoglutarate dehydrogenase mRNA levels in enterocytes and a simultaneous reduction in ROS levels ( P < 0.05). Moreover, AKG treatment increased the activities of the antioxidant enzymes and the efficiency of cellular respiration ( P < 0.05). AKG also regulated the mRNA levels of the target genes involved in antioxidant responses and xenobiotic detoxification in enterocytes. Increases in the protein levels of SOD1, SOD2, CAR, RXRα, and UCP2 and marked reductions in the expression levels of Nrf2 and Keap1 proteins ( P < 0.05) were observed after AKG administration in the H2O2-induced piglets. Our results indicated that AKG may protect against oxidative stress by activating CAR signaling and modulating the expression of key antioxidant-related targets, which improves cellular respiration and antioxidant capacity. The in vivo and in vitro effects of AKG suggest that it may prove to be useful in the reduction of oxidative stress in animal and human trials and subsequent prevention of gastrointestinal pathologies.
Collapse
Affiliation(s)
- Liuqin He
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences , Hunan Normal University , Changsha 410081 , PR China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, 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 , Changsha 410125 , PR China
- National Engineering Laboratory for Rice and Byproduct Deep Processing , Center South University of Forestry and Technology , Changsha 410004 , PR China
| | - Jian Wu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, 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 , Changsha 410125 , PR China
| | - Wenjie Tang
- Sichuan Academy of Animal Sciences , Animal Breeding and Genetics Key Laboratory of Sichuan Province , Chengdu 610066 , PR China
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, 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 , Changsha 410125 , PR China
| | - Qinlu Lin
- National Engineering Laboratory for Rice and Byproduct Deep Processing , Center South University of Forestry and Technology , Changsha 410004 , PR China
| | - Feijun Luo
- National Engineering Laboratory for Rice and Byproduct Deep Processing , Center South University of Forestry and Technology , Changsha 410004 , PR China
| | - Yulong 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 , Changsha 410081 , PR China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, 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 , Changsha 410125 , PR China
- Hunan Co-Innovation Center of Animal Production Safety , CICAPS , Changsha 410128 , PR China
| | - Tiejun Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, 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 , Changsha 410125 , PR China
- Hunan Co-Innovation Center of Animal Production Safety , CICAPS , Changsha 410128 , PR China
| |
Collapse
|
32
|
Transcriptional Profiling Suggests Extensive Metabolic Rewiring of Human and Mouse Macrophages during Early Interferon Alpha Responses. Mediators Inflamm 2018; 2018:5906819. [PMID: 30147442 PMCID: PMC6083555 DOI: 10.1155/2018/5906819] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/24/2018] [Indexed: 12/31/2022] Open
Abstract
Emerging evidence suggests that cellular metabolism plays a critical role in regulating immune activation. Alterations in energy and lipid and amino acid metabolism have been shown to contribute to type I interferon (IFN) responses in macrophages, but the relationship between metabolic reprogramming and the establishment of early antiviral function remains poorly defined. Here, we used transcriptional profiling datasets to develop global metabolic signatures associated with early IFN-α responses in two primary macrophage model systems: mouse bone marrow-derived macrophages (BMM) and human monocyte-derived macrophages (MDM). Short-term stimulation with IFN-α (<4 hours) was associated with significant metabolic rewiring, with >500 metabolic genes altered in mouse and human macrophage models. Pathway and network analysis identified alterations in genes associated with cellular bioenergetics, cellular oxidant status, cAMP/AMP and cGMP/GMP ratios, branched chain amino acid catabolism, cell membrane composition, fatty acid synthesis, and β-oxidation as key features of early IFN-α responses. These changes may have important implications for initial establishment of antiviral function in these cells.
Collapse
|
33
|
He L, Zhou X, Huang N, Li H, Cui Z, Tian J, Jiang Q, Liu S, Wu J, Li T, Yao K, Yin Y. Administration of alpha-ketoglutarate improves epithelial restitution under stress injury in early-weaning piglets. Oncotarget 2017; 8:91965-91978. [PMID: 29190890 PMCID: PMC5696156 DOI: 10.18632/oncotarget.20555] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/07/2017] [Indexed: 12/21/2022] Open
Abstract
Alpha-ketoglutarate (AKG) is an important cellular metabolite that participates in energy production and amino acid metabolism. However, the protective effects and mechanism of AKG on mucosal lesions have not been well understood. This study was conducted to investigate the effects of dietary AKG supplementation on epithelial restitution in early-weaning piglets under Escherichia coli lipopolysaccharide (LPS) induction. A total of 32 weaned piglets were used in a 2 × 2 factorial design; the major factors were dietary treatment (basal diet or AKG diet) and inflammatory challenge (LPS or saline). The results showed that AKG supplementation improved the growth performance and intestinal morphology in the LPS-induced early-weaning piglets. Compared with the basal diet, the AKG diet remarkably decreased the concentration and mRNA expression of intestinal inflammatory cytokines (IL-1β, IL-6, and IL-12) in the LPS-induced piglets. Moreover, AKG administration upregulated the mRNA expression of nutrient-sensing transporters (GLUT-2, SGLT-1, PEPT-1, I-FABP2) in the small intestine of both saline- and LPS-treated piglets, and improved the distribution and expression of tight-junction genes andproteins (ZO-1, Occludin, Claudins, E-cadherin). Collectively, our findings indicate that AKG has the potential to alleviate intestinal inflammatory response and improve epithelial restitution and nutrient-sensing ability under stress injury in early-weaning piglets, and it also provides an experimental basis for enteral use of AKG in swine production and clinical application to prevent intestinal epithelial damage.
Collapse
Affiliation(s)
- Liuqin He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan 410125, China.,University of Chinese Academy of Sciences, Beijing, 10039, China
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan 410125, China
| | - Niu Huang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Huan Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Zhijie Cui
- Xiangtan University, Xiangtan, Hunan 411105, China
| | - Junquan Tian
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan 410125, China.,University of Chinese Academy of Sciences, Beijing, 10039, China
| | - Qian Jiang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan 410125, China.,University of Chinese Academy of Sciences, Beijing, 10039, China
| | - Shaojuan Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan 410125, China.,University of Chinese Academy of Sciences, Beijing, 10039, China
| | - Jian Wu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan 410125, China.,University of Chinese Academy of Sciences, Beijing, 10039, China
| | - Tiejun Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan 410125, China.,Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, 410128, China
| | - Kang Yao
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan 410125, China.,Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, 410128, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan 410125, China.,Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, 410128, China.,Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410006, China
| |
Collapse
|
34
|
He L, Zhou X, Huang N, Li H, Li T, Yao K, Tian Y, Hu CAA, Yin Y. Functions of pregnane X receptor in self-detoxification. Amino Acids 2017; 49:1999-2007. [PMID: 28534176 DOI: 10.1007/s00726-017-2435-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/03/2017] [Indexed: 12/19/2022]
Abstract
Pregnane X receptor (PXR, NR1I2), a member of the nuclear receptor superfamily, is a crucial regulator of nutrient metabolism and metabolic detoxification such as metabolic syndrome, xenobiotic metabolism, inflammatory responses, glucose, cholesterol and lipid metabolism, and endocrine homeostasis. Notably, much experimental and clinical evidence show that PXR senses xenobiotics and triggers the detoxification response to prevent diseases such as diabetes, obesity, intestinal inflammatory diseases and liver fibrosis. In this review we summarize recent advances on remarkable metabolic and regulatory versatility of PXR, and we emphasizes its role and potential implication as an effective modulator of self-detoxification in animals and humans.
Collapse
Affiliation(s)
- Liuqin He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock and Poultry, Changsha, 410125, Hunan, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock and Poultry, Changsha, 410125, Hunan, China
| | - Niu Huang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China
| | - Huan Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China
| | - Tiejun Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock and Poultry, Changsha, 410125, Hunan, China.,Hunan Co-Innovation Center of Animal Production Safety, Changsha, 410128, Hunan, China
| | - Kang Yao
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock and Poultry, Changsha, 410125, Hunan, China. .,College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China. .,Hunan Co-Innovation Center of Animal Production Safety, Changsha, 410128, Hunan, China.
| | - Yanan Tian
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China.,Department of Veterinary Physiology and Pharmacology, Texas A & M University, College Station, TX, 77843, USA
| | - Chien-An Andy Hu
- Department of Biochemistry and Molecular Biology, University of New Mexico, Health Sciences Center, MSC08 4670, Albuquerque, USA
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock and Poultry, Changsha, 410125, Hunan, China. .,Hunan Co-Innovation Center of Animal Production Safety, Changsha, 410128, Hunan, China.
| |
Collapse
|