1
|
Cao Q, Fan J, Zou J, Wang W. Multi-omics analysis identifies BCAT2 as a potential pan-cancer biomarker for tumor progression and immune microenvironment modulation. Sci Rep 2024; 14:23371. [PMID: 39375392 PMCID: PMC11458862 DOI: 10.1038/s41598-024-74441-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024] Open
Abstract
Branched-chain amino acid transaminase 2 (BCAT2) encodes a crucial protein involved in the initial catalysis of branched-chain amino acid (BCAA) catabolism, with emerging evidence suggesting its association with tumor progression. This study explores BCAT2 in a pan-cancer multi-omics context and evaluates its prognostic significance. We utilized a multi-database approach, analyzing cBioPortal for genetic alterations, RNA-Seq data from TCGA and GTEx for expression patterns, and RSEM for transcript analysis. Protein expression and interaction networks were assessed using the Human Protein Atlas, UniProt, and STRING. Prognostic value was determined through Cox regression analysis of TCGA clinical survival data, while immune cell infiltration across various cancers was examined using TCGA data and the TIMER2 platform. Our results revealed that BCAT2 alterations are primarily amplifications and is upregulated in various tumors, correlating with poor survival rates in several tumor types, including GBMLGG, LGG, and UVM. Elevated BCAT2 protein levels were common in pan-cancer, interacting with a range of metabolic enzymes. Additionally, BCAT2 expression significantly influenced CD4+ T cells, CD8+ T cells, and Treg cells infiltration, with varied correlations across cancer types. These findings indicate BCAT2 as a potential biomarker for cancer diagnosis and therapy, potentially regulating key metabolic and immune factors to mediate tumor progression and the microenvironment.
Collapse
Affiliation(s)
- Qixuan Cao
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Jie Fan
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Jian Zou
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| | - Wei Wang
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
2
|
Ma Q, Li H, Song Z, Deng Z, Huang W, Liu Q. Fueling the fight against cancer: Exploring the impact of branched-chain amino acid catalyzation on cancer and cancer immune microenvironment. Metabolism 2024; 161:156016. [PMID: 39222743 DOI: 10.1016/j.metabol.2024.156016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Metabolism of Branched-chain amino acids (BCAAs) is essential for the nutrient necessities in mammals. Catalytic enzymes serve to direct the whole-body BCAAs oxidation which involve in the development of various metabolic disorders. The reprogrammed metabolic elements are also responsible for malignant oncogenic processes, and favor the formation of distinctive immunosuppressive microenvironment surrounding different cancers. The impotent immune surveillance related to BCAAs dysfunction is a novel topic to investigate. Here we focus on the BCAA catalysts that contribute to metabolic changes and dysregulated immune reactions in cancer progression. We summarize the current knowledge of BCAA catalyzation, highlighting the interesting roles of BCAA metabolism in the treatment of cancers.
Collapse
Affiliation(s)
- Qianquan Ma
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, China
| | - Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province
| | - Zhihao Song
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province.
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province.
| |
Collapse
|
3
|
Chamroonkul N, Rujeerapaiboon N, Sripongpun P, Kaewdech A, Piratvisuth T. The efficacy of branched-chain amino acid granules to restore phagocytic activity in cirrhosis patients, a randomized controlled trial. Front Nutr 2023; 10:1142206. [PMID: 37252239 PMCID: PMC10213217 DOI: 10.3389/fnut.2023.1142206] [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: 01/11/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
Background Infection is a detrimental complication among cirrhotic patients, leading to major morbidity and mortality. Reduction in phagocytic activation, as part of immunoparesis, is a distinctive key component of cirrhosis-associated immune dysfunction (CAID) and predicts the development of infection. However, there are limited data on immunotherapeutic approaches to restore phagocytosis. Aims We aimed to determine the effect of branched-chain amino acid (BCAA) granules on phagocytic activity in patients with CAID. Methods In this double-blind randomized controlled trial, Participants were randomly assigned (1:1 ratio stratified by Child-Pugh status) to receive either BCAA granules or placebo. In the 3rd and 6th months, phagocytic activity was assessed by flow cytometry. The primary endpoint was the restoration of innate immunity at the 6th month, defined as ≥75% phagocytic activity; the secondary endpoints were the accretion of phagocytic activity and hospitalization due to infection. Results A total of 37 patients were included. There were no differences among the patients in the baseline characteristics and phagocytic activity. At the 6th month, a higher proportion of patients with phagocytic restoration was observed in the BCAA granule group compared to the placebo group (68 vs. 5.6%, p < 0.001). The mean phagocytic activity was 75.4 and 63.4% in the BCAA granule and placebo groups, respectively (p < 0.001). Progressive accretion of phagocytic activity was observed during the 3rd and 6th months. There was no difference in hospitalization due to infection (3 vs. 2 events, p = 0.487). Conclusion Our results suggest that BCAA granules significantly restore phagocytic activity across various stages of cirrhosis. A longer follow-up period is required to demonstrate infection prevention.Clinical Trial Registration: www.clinicaltrials.in.th, TCTR20190830005.
Collapse
Affiliation(s)
- Naichaya Chamroonkul
- Gastroenterology and Hepatology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Natthapat Rujeerapaiboon
- Gastroenterology and Hepatology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
- Gastroenterology Endoscopy and Motility Center, Ramathibodi Hospital, Mahidol University, Ratchathewi, Thailand
| | - Pimsiri Sripongpun
- Gastroenterology and Hepatology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Apichat Kaewdech
- Gastroenterology and Hepatology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Teerha Piratvisuth
- NKC Institute of Gastroenterology and Hepatology, Songklanagarind Hospital, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
4
|
Cheng Z, Yang L, Chu H. The Gut Microbiota: A Novel Player in Autoimmune Hepatitis. Front Cell Infect Microbiol 2022; 12:947382. [PMID: 35899041 PMCID: PMC9310656 DOI: 10.3389/fcimb.2022.947382] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Autoimmune hepatitis (AIH) is a chronic immune-mediated liver disease distributed globally in all ethnicities with increasing prevalence. If left untreated, the disease will lead to cirrhosis, liver failure, or death. The intestinal microbiota is a complex ecosystem located in the human intestine, which extensively affects the human physiological and pathological processes. With more and more in-depth understandings of intestinal microbiota, a substantial body of studies have verified that the intestinal microbiota plays a crucial role in a variety of digestive system diseases, including alcohol-associated liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD). However, only a few studies have paid attention to evaluate the relationship between AIH and the intestinal microbiota. While AIH pathogenesis is not fully elucidated yet, some studies have indicated that intestinal microbiota putatively made significant contributions to the occurrence and the development of AIH by triggering several specific signaling pathways, altering the metabolism of intestinal microbiota, as well as modulating the immune response in the intestine and liver. By collecting the latest related literatures, this review summarized the increasing trend of the aerobic bacteria abundance in both AIH patients and AIH mice models. Moreover, the combination of specific bacteria species was found distinct to AIH patients, which could be a promising tool for diagnosing AIH. In addition, there were alterations of luminal metabolites and immune responses, including decreased short-chain fatty acids (SCFAs), increased pathogen associated molecular patterns (PAMPs), imbalanced regulatory T (Treg)/Th17 cells, follicular regulatory T (TFR)/follicular helper T (TFH) cells, and activated natural killer T (NKT) cells. These alterations participate in the onset and the progression of AIH via multiple mechanisms. Therefore, some therapeutic methods based on restoration of intestinal microbiota composition, including probiotics and fecal microbiota transplantation (FMT), as well as targeted intestinal microbiota-associated signaling pathways, confer novel insights into the treatment for AIH patients.
Collapse
Affiliation(s)
| | - Ling Yang
- *Correspondence: Huikuan Chu, ; Ling Yang, ;
| | - Huikuan Chu
- *Correspondence: Huikuan Chu, ; Ling Yang, ;
| |
Collapse
|
5
|
Yan Y, Chen J, Liang Q, Zheng H, Ye Y, Nan W, Zhang X, Gao H, Li Y. Metabolomics profile in acute respiratory distress syndrome by nuclear magnetic resonance spectroscopy in patients with community-acquired pneumonia. Respir Res 2022; 23:172. [PMID: 35761396 PMCID: PMC9235271 DOI: 10.1186/s12931-022-02075-w] [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: 12/13/2021] [Accepted: 05/23/2022] [Indexed: 11/20/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is a challenging clinical problem. Discovering the potential metabolic alterations underlying the ARDS is important to identify novel therapeutic target and improve the prognosis. Serum and urine metabolites can reflect systemic and local changes and could help understanding metabolic characterization of community-acquired pneumonia (CAP) with ARDS. Methods Clinical data of patients with suspected CAP at the First Affiliated Hospital of Wenzhou Medical University were collected from May 2020 to February 2021. Consecutive patients with CAP were enrolled and divided into two groups: CAP with and without ARDS groups. 1H nuclear magnetic resonance-based metabolomics analyses of serum and urine samples were performed before and after treatment in CAP with ARDS (n = 43) and CAP without ARDS (n = 45) groups. Differences metabolites were identifed in CAP with ARDS. Furthermore, the receiver operating characteristic (ROC) curve was utilized to identify panels of significant metabolites for evaluating therapeutic effects on CAP with ARDS. The correlation heatmap was analyzed to further display the relationship between metabolites and clinical characteristics. Results A total of 20 and 42 metabolites were identified in the serum and urine samples, respectively. Serum metabolic changes were mainly involved in energy, lipid, and amino acid metabolisms, while urine metabolic changes were mainly involved in energy metabolism. Elevated levels of serum 3-hydroxybutyrate, lactate, acetone, acetoacetate, and decreased levels of serum leucine, choline, and urine creatine and creatinine were detected in CAP with ARDS relative to CAP without ARDS. Serum metabolites 3-hydroxybutyrate, acetone, acetoacetate, citrate, choline and urine metabolite 1-methylnicotinamide were identified as a potential biomarkers for assessing therapeutic effects on CAP with ARDS, and with AUCs of 0.866 and 0.795, respectively. Moreover, the ROC curve analysis revealed that combined characteristic serum and urine metabolites exhibited a better classification system for assessing therapeutic effects on CAP with ARDS, with a AUC value of 0.952. In addition, differential metabolites strongly correlated with clinical parameters in patients with CAP with ARDS. Conclusions Serum- and urine-based metabolomics analyses identified characteristic metabolic alterations in CAP with ARDS and might provide promising circulatory markers for evaluating therapeutic effects on CAP with ARDS. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02075-w.
Collapse
Affiliation(s)
- Yongqin Yan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, China
| | - Jianuo Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, China
| | - Qian Liang
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hong Zheng
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yiru Ye
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, China
| | - Wengang Nan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, China
| | - Xi Zhang
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongchang Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, China. .,Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Yuping Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, China.
| |
Collapse
|
6
|
Effects of Inclusion of N-Carbamylglutamate in the Non-Protein Diet on Growth and Slaughter Performance, Meat Quality, Nitrogen Metabolism and Antioxidant of Holstein Bulls. Animals (Basel) 2021; 12:ani12010033. [PMID: 35011139 PMCID: PMC8749524 DOI: 10.3390/ani12010033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Using dietary non-protein nitrogen is an effective way to decrease the dependence on protein resources in cattle production. N-carbamylglutamate (NCG) is a structural analogue of N-acetylglutamate (NAG), which is a precursor of endogenous Arg synthesis. NCG improves urea cycling and enhances the endogenous synthesis of Arg, nitric oxide synthase and NO. The present study showed that beef benefited from being fed NCG product in the urea diet by enhancing its growth and slaughter performance, meat quality, nitrogen metabolism and plasma amino acids. Abstract The objectives of this experiment were to investigate the effects of N-carbamylglutamate (NCG) on growth and slaughter performance, meat quality, nitrogen utilization, plasma antioxidant and amino acids of Holstein bulls. In this case, 24 Holstein bulls (490 ± 29.0 kg of body weights and 540 ± 6.1 d of age) were blocked by body weights and age and randomly assigned to 1 of 4 groups: (1) CON group: bulls were fed the control diet, (2) CON + NCG group: bulls were fed the control diet with 40 mg/kg BW NCG, (3) Urea group: bulls were fed the urea diet, and (4) Urea + NCG group: bulls were fed the urea diet with 40 mg/kg BW NCG. Feeding NCG significantly improved ADG, FCR, DM and CP digestibility, carcass weight, slaughter weight, DOP, eye muscle area, shear force (p = 0.001) and reduced L* of color, drip loss and cooking loss. Concurrently, feeding the urea diet induced a decreased ADG, carcass weight and slaughter weight, DOP, eye muscle area and shear force. NCG decreased contents of fecal N and urinary N, plasma urea in bulls and ammonia but increased N retention and utilization, plasma NO, plasma Arg, Leu, Ile and Tyr. On the other hand, feeding the urea diet increased urinary N, plasma urea and ammonia. Thus the study efficiently demonstrates that beef benefited from being fed a NCG product in the urea diet by enhancing its growth and slaughter performance, meat quality, nitrogen metabolism and plasma amino acids.
Collapse
|
7
|
Aquilani R, Zuccarelli GC, Maestri R, Boselli M, Dossena M, Baldissarro E, Boschi F, Buonocore D, Verri M. Essential amino acid supplementation is associated with reduced serum C-reactive protein levels and improved circulating lymphocytes in post-acute inflamed elderly patients. Int J Immunopathol Pharmacol 2021; 35:20587384211036823. [PMID: 34387512 PMCID: PMC8366127 DOI: 10.1177/20587384211036823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Persistent systemic inflammation leads to multidistrectual body dysfunctions. Attenuation of inflammation may improve patients’ functional and life prognoses. We hypothesized that essential amino acids (EAAs) given to elderly patients in rehabilitation after acute diseases may be associated with a reduced inflammatory state. Therefore, this retrospective study investigated whether the supplementation of EAAs – modulators of immune competence – was associated with a reduced inflammation rate in elderly patients. Methods The medical records of 282 patients admitted to the rehabilitation (rehab) institute after acute index events (surgery or medical diseases) (age: 81.18 ± 8.58 years; females: 67.9%) were analyzed. Results 46 patients (16.3% of the entire population) had received EAA supplements (S), whereas the remaining 236 patients had not (N-S). Systemic inflammation (I) (serum C-reactive protein (CRP) > 0.5 mg/dL) was present in 67.4% of the I-S group and 57.2% of the I-N-S group. During rehab, the I-S group (but not the I-N-S group) showed a reduction in CRP levels (p = 0.03) and an increase in circulating lymphocytes (p = 0.035), immune cells of the adaptive immune system. C-reactive protein levels remained virtually unchanged in non-inflamed patients who received supplements but increased in non-inflamed patients who did not receive supplements (p = 0.05). Stratified for developed infections, CRP levels reduced in S patients (p = 0.008) but did not in N-S patients. Conclusion EAA supplementation was associated with reduced inflammation in both inflamed and infected patients. In addition, EAA supplementation was associated with increased circulating lymphocytes in inflamed patients.
Collapse
Affiliation(s)
- Roberto Aquilani
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Ginetto C Zuccarelli
- Geriatric Institute P. Redaelli - Reparti di Riabilitazione Geriatrica e di Mantenimento, Vimodrone (Milano), Italy
| | - Roberto Maestri
- Department of Biomedical Engineering of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano (PV), Italy
| | - Mirella Boselli
- Neurorehabilitation Unit of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano (PV), Italy
| | - Maurizia Dossena
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Eleonora Baldissarro
- Complex Structure of Recovery and Functional Re-education - ASL 3, Genova, Italy
| | - Federica Boschi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Daniela Buonocore
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Manuela Verri
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
8
|
Bai R, Bai M, Zhao Z, Chen N, Yang Y, Tang Z. Alternation of supragingival microbiome in patients with cirrhosis of different Child-Pugh scores. Oral Dis 2020; 28:233-242. [PMID: 33274586 DOI: 10.1111/odi.13743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/11/2020] [Accepted: 11/29/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The aim of this study was to analyze the differences in the taxonomy and functions of oral microbiome between patients with and without cirrhosis. MATERIALS AND METHODS In this study, V4-16S rDNA amplicon sequencing was used to compare the difference of supragingival microbiome in 42 patients and 12 healthy individuals. RESULTS Overall, 3,223,529 clean reads were generated, with an average of 59,694 ± 1,548 clean reads per sample. A total of 30 phyla, 78 classes, 116 orders, 167 families, 228 genera, and 114 species were detected in the 54 samples. The differences were detected among groups at each taxonomical level. Functional prediction showed that patients with cirrhosis had a significant higher proportion of the genes associated with carbohydrate transport and metabolism, defense mechanisms, infectious diseases, membrane transport, etc. compared with healthy individuals (p < .05). CONCLUSIONS In conclusion, significant differences were observed in compositions and predictive functions of the supragingival microbiome between patients with cirrhosis and that in healthy people. These findings will provide a new insight into the understanding of pathogenesis, diagnosis, prognosis, and therapy of cirrhosis.
Collapse
Affiliation(s)
- Rushui Bai
- Hunan Key Laboratory of Oral Health Research, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, China.,Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Mingshan Bai
- Seventh Ward, Tangshan Infectious Disease Hospital, Tangshan, China
| | - Zijia Zhao
- Hunan Key Laboratory of Oral Health Research, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, China
| | - Ningxin Chen
- Hunan Key Laboratory of Oral Health Research, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, China
| | - Yan Yang
- Hunan Key Laboratory of Oral Health Research, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, China
| | - Zhangui Tang
- Hunan Key Laboratory of Oral Health Research, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
9
|
Shen J, Xiang S, Peng M, Zhou Z, Wu Z. Mechanisms of Resistance to Schistosoma japonicum Infection in Microtus fortis, the Natural Non-permissive Host. Front Microbiol 2020; 11:2092. [PMID: 33013763 PMCID: PMC7494751 DOI: 10.3389/fmicb.2020.02092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
Human schistosomiasis, which is caused by schistosomes, is a zoonosis that is difficult to control because of the many reservoir hosts. However, Microtus fortis is the only mammal that is naturally resistant to Schistosoma japonicum infection known in China, in which S. japonicum growth and development were arrested on day 12, and the worms eliminated on day 20 post-infection. In this review, we present an overview of the established and purported mechanisms of resistance to S. japonicum infection in M. fortis in comparison to Rattus norvegicus, a semi-permissive host. Clarifying the mechanism of this efficient resistance can help us to better understand host-parasite interaction and to provide better methods to control schistosomiasis.
Collapse
Affiliation(s)
- Jia Shen
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Suoyu Xiang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Mei Peng
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Zhijun Zhou
- Department of Laboratory Animal Science, Xiangya Medical College, Central South University, Changsha, China.,Hunan Key Laboratory of Animal Models for Human Diseases, Changsha, China
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| |
Collapse
|
10
|
Coleman DN, Lopreiato V, Alharthi A, Loor JJ. Amino acids and the regulation of oxidative stress and immune function in dairy cattle. J Anim Sci 2020; 98:S175-S193. [PMID: 32810243 PMCID: PMC7433927 DOI: 10.1093/jas/skaa138] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Affiliation(s)
| | - Vincenzo Lopreiato
- Department of Health Science, Interdepartmental Services Centre of Veterinary for Human and Animal Health, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Abdulrahman Alharthi
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Juan J Loor
- Department of Animal Sciences, University of Illinois, Urbana, IL.,Division of Nutritional Sciences, University of Illinois, Urbana, IL
| |
Collapse
|
11
|
Abstract
Cirrhosis is a multisystemic disease wherein inflammatory responses originating from advanced liver disease and its sequelae affect distant compartments. Patients with cirrhosis are susceptible to bacterial infections, which may precipitate acute decompensation and acute-on-chronic liver failure, both of which are associated with high short-term mortality. Innate immune cells are an essential first line of defence against pathogens. Activation of liver macrophages (Kupffer cells) and resident mastocytes generate proinflammatory and vaso-permeating mediators that induce accumulation of neutrophils, lymphocytes, eosinophils and monocytes in the liver, and promote tissue damage. During cirrhosis progression, damage- and pathogen-associated molecular patterns activate immune cells and promote development of systemic inflammatory responses which may involve different tissues and compartments. The antibacterial function of circulating neutrophils and monocytes is gradually and severely impaired as cirrhosis worsens, contributing to disease progression. The mechanisms underlying impaired antimicrobial responses are complex and incompletely understood. This review focuses on the continuous and distinct perturbations arising in innate immune cells during cirrhosis, including their impact on disease progression, as well as reviewing potential therapeutic targets.
Collapse
|
12
|
Gu C, Mao X, Chen D, Yu B, Yang Q. Isoleucine Plays an Important Role for Maintaining Immune Function. Curr Protein Pept Sci 2019; 20:644-651. [PMID: 30843485 DOI: 10.2174/1389203720666190305163135] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/30/2018] [Accepted: 01/10/2019] [Indexed: 12/16/2022]
Abstract
Branched chain amino acids are the essential nutrients for humans and many animals. As functional amino acids, they play important roles in physiological functions, including immune functions. Isoleucine, as one of the branched chain amino acids, is also critical in physiological functions of the whole body, such as growth, immunity, protein metabolism, fatty acid metabolism and glucose transportation. Isoleucine can improve the immune system, including immune organs, cells and reactive substances. Recent studies have also shown that isoleucine may induce the expression of host defense peptides (i.e., β-defensins) that can regulate host innate and adaptive immunity. In addition, isoleucine administration can restore the effect of some pathogens on the health of humans and animals via increasing the expression of β-defensins. Therefore, the present review will emphatically discuss the effect of isoleucine on immunity while summarizing the relationship between branched chain amino acids and immune functions.
Collapse
Affiliation(s)
- Changsong Gu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease- Resistance Nutrition, Chinese Ministry of Education, Chengdu, China
| | - Xiangbing Mao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease- Resistance Nutrition, Chinese Ministry of Education, Chengdu, China
| | - Daiwen Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease- Resistance Nutrition, Chinese Ministry of Education, Chengdu, China
| | - Bing Yu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease- Resistance Nutrition, Chinese Ministry of Education, Chengdu, China
| | - Qing Yang
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, KS, United States
| |
Collapse
|
13
|
van Sadelhoff JHJ, Perez Pardo P, Wu J, Garssen J, van Bergenhenegouwen J, Hogenkamp A, Hartog A, Kraneveld AD. The Gut-Immune-Brain Axis in Autism Spectrum Disorders; A Focus on Amino Acids. Front Endocrinol (Lausanne) 2019; 10:247. [PMID: 31057483 PMCID: PMC6477881 DOI: 10.3389/fendo.2019.00247] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/29/2019] [Indexed: 12/25/2022] Open
Abstract
Autism spectrum disorder (ASD) is a range of neurodevelopmental conditions that affect communication and social behavior. Besides social deficits, systemic inflammation, gastrointestinal immune-related problems, and changes in the gut microbiota composition are characteristic for people with ASD. Animal models showed that these characteristics can induce ASD-associated behavior, suggesting an intimate relationship between the microbiota, gut, immune system and the brain in ASD. Multiple factors can contribute to the development of ASD, but mutations leading to enhanced activation of the mammalian target of rapamycin (mTOR) are reported frequently. Hyperactivation of mTOR leads to deficits in the communication between neurons in the brain and to immune impairments. Hence, mTOR might be a critical factor linking the gut-brain-immune axis in ASD. Pharmacological inhibition of mTOR is shown to improve ASD-associated behavior and immune functions, however, the clinical use is limited due to severe side reactions. Interestingly, studies have shown that mTOR activation can also be modified by nutritional stimuli, in particular by amino acids. Moreover, specific amino acids are demonstrated to inhibit inflammation, improve gut barrier function and to modify the microbiota composition. In this review we will discuss the gut-brain-immune axis in ASD and explore the potential of amino acids as a treatment option for ASD, either via modification of mTOR activity, the immune system or the gut microbiota composition.
Collapse
Affiliation(s)
- Joris H. J. van Sadelhoff
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Paula Perez Pardo
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jiangbo Wu
- Laboratory of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Astrid Hogenkamp
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Anita Hartog
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Veterinary Pharmacology, Institute for Risk Assessment Studies, Faculty of Veterinary Sciences, Utrecht University, Utrecht, Netherlands
- *Correspondence: Aletta D. Kraneveld
| |
Collapse
|
14
|
The Value of the C-Reactive Protein-to-Albumin Ratio is Useful for Predicting Survival of Patients with Child-Pugh Class A Undergoing Liver Resection for Hepatocellular Carcinoma. World J Surg 2018; 42:2218-2226. [PMID: 29288307 DOI: 10.1007/s00268-017-4446-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Although a recent study has shown that the C-reactive protein-to-albumin ratio (CAR) can predict the survival in patients with hepatocellular carcinoma (HCC), it is unclear whether CAR can predict the survival after surgery. OBJECTIVE To investigate the utility of CAR for prediction of postoperative survival among HCC patients with Child-Pugh class A. METHODS We retrospectively reviewed 239 patients with Child-Pugh class A who were newly diagnosed with HCC and received initial liver resection. Univariate and multivariate analyses using the Cox proportional hazard model were performed to detect clinical characteristics that correlated with overall survival (OS), and their cutoff values were identified using receiver operating characteristic curve analyses. The cutoff value of CAR was 0.028. Kaplan-Meier analysis and the log-rank test were used for the comparison of OS and disease-free survival (DFS) between two CAR groups (>0.028/≤0.028). RESULTS Multivariate analysis using 16 clinical characteristics selected by univariate analyses revealed that CAR (>0.028/≤0.028) (HR, 3.211; 95% CI 1.065-9.680; P = 0.038) was significantly associated with OS, as well as anatomical resection (presence/absence) (HR, 0.275; 95% CI 0.119-0.635; P = 0.275). A significant difference in OS and DFS was observed between patients with low CAR (≤0.028) and patients with high CAR (>0.028). CONCLUSIONS CAR is a useful predictor of postoperative survival among HCC patients with Child-Pugh class A.
Collapse
|
15
|
Nie C, He T, Zhang W, Zhang G, Ma X. Branched Chain Amino Acids: Beyond Nutrition Metabolism. Int J Mol Sci 2018; 19:E954. [PMID: 29570613 PMCID: PMC5979320 DOI: 10.3390/ijms19040954] [Citation(s) in RCA: 383] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/01/2018] [Accepted: 03/14/2018] [Indexed: 12/14/2022] Open
Abstract
Branched chain amino acids (BCAAs), including leucine (Leu), isoleucine (Ile), and valine (Val), play critical roles in the regulation of energy homeostasis, nutrition metabolism, gut health, immunity and disease in humans and animals. As the most abundant of essential amino acids (EAAs), BCAAs are not only the substrates for synthesis of nitrogenous compounds, they also serve as signaling molecules regulating metabolism of glucose, lipid, and protein synthesis, intestinal health, and immunity via special signaling network, especially phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signal pathway. Current evidence supports BCAAs and their derivatives as the potential biomarkers of diseases such as insulin resistance (IR), type 2 diabetes mellitus (T2DM), cancer, and cardiovascular diseases (CVDs). These diseases are closely associated with catabolism and balance of BCAAs. Hence, optimizing dietary BCAA levels should have a positive effect on the parameters associated with health and diseases. This review focuses on recent findings of BCAAs in metabolic pathways and regulation, and underlying the relationship of BCAAs to related disease processes.
Collapse
Affiliation(s)
- Cunxi Nie
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing 100193, China.
- College of Animal Science and Technology, Shihezi University, No. 221. Beisi Road, Shihezi, Xinjiang 832003, China.
| | - Ting He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing 100193, China.
| | - Wenju Zhang
- College of Animal Science and Technology, Shihezi University, No. 221. Beisi Road, Shihezi, Xinjiang 832003, China.
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078, USA.
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing 100193, China.
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
16
|
Tajiri K, Shimizu Y. Branched-chain amino acids in liver diseases. Transl Gastroenterol Hepatol 2018; 3:47. [PMID: 30148232 PMCID: PMC6088198 DOI: 10.21037/tgh.2018.07.06] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/06/2018] [Indexed: 02/05/2023] Open
Abstract
Branched chain amino acids (BCAAs) are involved in various bioprocess such as protein metabolism, gene expression, insulin resistance and proliferation of hepatocytes. BCAAs have also been reported to suppress the growth of hepatocellular carcinoma (HCC) cells in vitro and to be required for immune cells to perform the function. In advanced cirrhotic patients, it has been clarified that serum concentrations of BCAA are decreased, whereas those of aromatic amino acids (AAAs) are increased. These alterations are thought to be the causes of hepatic encephalopathy (HE), sarcopenia and hepatocarcinogenesis and may be associated with the poor prognosis of patients with these conditions. Administration of BCAA-rich medicines has shown positive results in patients with cirrhosis.
Collapse
Affiliation(s)
- Kazuto Tajiri
- Department of Gastroenterology, Toyama University Hospital, Toyama, Japan
| | | |
Collapse
|
17
|
Zhou H, Yu B, Gao J, Htoo JK, Chen D. Regulation of intestinal health by branched-chain amino acids. Anim Sci J 2017; 89:3-11. [PMID: 29164733 DOI: 10.1111/asj.12937] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/11/2017] [Indexed: 12/15/2022]
Abstract
Besides its primary role in the digestion and absorption of nutrients, the intestine also interacts with a complex external milieu, and is the first defense line against noxious pathogens and antigens. Dysfunction of the intestinal barrier is associated with enhanced intestinal permeability and development of various gastrointestinal diseases. The branched-chain amino acids (BCAAs) are important nutrients, which are the essential substrates for protein biosynthesis. Recently, emerging evidence showed that BCAAs are involved in maintaining intestinal barrier function. It has been reported that dietary supplementation with BCAAs promotes intestinal development, enhances enterocyte proliferation, increases intestinal absorption of amino acids (AA) and glucose, and improves the immune defenses of piglets. The underlying mechanism of these effects is mediated by regulating expression of genes and proteins associate with various signaling pathways. In addition, BCAAs promote the production of beneficial bacteria in the intestine of mice. Compelling evidence supports the notion that BCAAs play important roles in both nutrition and intestinal health. Therefore, as functional amino acids with various physiological effects, BCAAs hold key roles in promoting intestinal development and health in animals and humans.
Collapse
Affiliation(s)
- Hua Zhou
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, China
| | - Jun Gao
- Evonik Degussa (China) Co. Ltd., Beijing, China
| | | | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, China
| |
Collapse
|
18
|
Dietary and metabolic modulators of hepatic immunity. Semin Immunopathol 2017; 40:175-188. [PMID: 29110070 DOI: 10.1007/s00281-017-0659-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022]
Abstract
The liver is the central metabolic organ of the organism and is thus constantly exposed to gut-derived dietary and microbial antigens. The liver maintains homoeostatic tolerance to these mostly harmless antigens. However, the liver also functions as a barrier organ to harmful pathogens and is thus permissive to liver inflammation. The regulation of the delicate balance between liver tolerance and liver inflammation is of vital importance for the organism. In recent years, a general role for dietary components and metabolites as immune mediators has been emerging. However, although the liver is exposed to a great deal of metabolic mediators, surprisingly, little is known about their actual role in the regulation of hepatic immune responses. Here, we will explore the possible impacts of metabolic mediators for homoeostatic and pathological immunity in the liver, by highlighting selected examples of metabolic immune regulation in the liver.
Collapse
|
19
|
Noceti O, Pouché L, Esperón P, Lens D, Vital M, Touriño C, Gerona S, Woillard JB, Marquet P. Activity of the Calcineurin Pathway in Patients on the Liver Transplantation Waiting List: Factors of Variability and Response to Tacrolimus Inhibition. Clin Chem 2017; 63:1734-1744. [DOI: 10.1373/clinchem.2017.272534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 08/02/2017] [Indexed: 01/10/2023]
Abstract
Abstract
BACKGROUND
We sought to evaluate, in patients on a liver transplantation waiting list, potential biomarkers of the base calcineurin pathway activity with use of a new model of nonstimulated peripheral blood mononuclear cells (PBMC) and ex vivo response to tacrolimus (TAC).
METHODS
The calcineurin pathway activity was explored ex vivo in stimulated and nonstimulated PBMC from 19 patients. The inhibition of NFAT1 translocation to PBMC nuclei, expression of intracellular IL-2, and membrane CD25 in different T-cell subsets were measured by multiparametric flow cytometry before and after exposure to TAC. We also studied the influence on the individual response of polymorphisms in 3 key genes of the calcineurin pathway: PPIA, PPP3CA, and IL2RA.
RESULTS
All pharmacodynamics profiles closely fitted an I/Imax sigmoid model. Interindividual variability was higher in nonstimulated than in stimulated conditions, as well as in the presence of TAC. IL-2+CD8+ cells at TAC Imax showed the highest interindividual variability, suggesting its usefulness as a biomarker of individual TAC effects integrating many different sources of regulation and variability. Moreover, in the absence of TAC, patients with end-stage liver disease exhibited lower NFAT1 translocation and T-cell activation than healthy volunteers from a previous study under similar conditions. Multivariate statistical analysis showed strong and significant associations between TAC pharmacodynamic parameters and 2 polymorphisms in the gene-coding cyclophilin A (rs8177826 and rs6850).
CONCLUSIONS
We show the feasibility of using nonstimulated PBMCs to explore the calcineurin pathway under more physiologic conditions and point toward potential biomarkers for TAC pharmacodynamic monitoring. ClinicalTrials.gov Identifier: NCT01760356
Collapse
Affiliation(s)
- Ofelia Noceti
- U850 INSERM, University of Limoges, CHU Limoges, FHU SUPORT, Limoges, France
- Clinical Biochemistry Department, School of Chemistry, Universidad de la República, Montevideo, Uruguay
- Liver Diseases Department, National Center for Liver Transplantation, Hospital Central de las Fuerzas Armadas, Montevideo, Uruguay
| | - Lucie Pouché
- U850 INSERM, University of Limoges, CHU Limoges, FHU SUPORT, Limoges, France
| | - Patricia Esperón
- Clinical Biochemistry Department, School of Chemistry, Universidad de la República, Montevideo, Uruguay
| | - Daniela Lens
- Department of Fundamental Medicine, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Marcelo Vital
- Clinical Biochemistry Department, School of Chemistry, Universidad de la República, Montevideo, Uruguay
| | - Cristina Touriño
- Department of Fundamental Medicine, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Solange Gerona
- Liver Diseases Department, National Center for Liver Transplantation, Hospital Central de las Fuerzas Armadas, Montevideo, Uruguay
| | | | - Pierre Marquet
- U850 INSERM, University of Limoges, CHU Limoges, FHU SUPORT, Limoges, France
| |
Collapse
|
20
|
High throughput data analyses of the immune characteristics of Microtus fortis infected with Schistosoma japonicum. Sci Rep 2017; 7:11311. [PMID: 28900150 PMCID: PMC5595801 DOI: 10.1038/s41598-017-11532-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 08/11/2017] [Indexed: 12/11/2022] Open
Abstract
Microtus fortis exhibits natural resistance against Schistosoma japonicum, and the parasite cannot grow and develop in M. fortis. Extensive research has been carried out, however, the associated mechanism remains unclear. In the present study, we analysed the combined data obtained from a cytokine chip assay, transcriptome, and metabolome. The cytokine profile from C57BL/6 and M. fortis mice was assessed before and after infection. Several cytokines increased during the second and third week post-infection. Some transcripts related to cytokine genes and associated proteins were also highly expressed (i.e., Hgf, C3, and Lbp). The liver metabolism of M. fortis following infection with S. japonicum was assessed. We identified 25 different metabolites between the uninfected and infected M. fortis, and 22 different metabolites between infected M. fortis and C57BL/6 mice. The metabolomic pathways of these differential metabolites were then analysed with MetPA, revealing that they were involved in histidine metabolism, valine, leucine, and isoleucine biosyntheses, and lysine degradation. Thus, the elevated expression of these metabolites and pathways may promote the phagocytic function of the neutrophils and natural killer cell activity following TLR activation. These results provide novel insight into the resistance mechanism of M. fortis against S. japonicum.
Collapse
|
21
|
Zhang S, Zeng X, Ren M, Mao X, Qiao S. Novel metabolic and physiological functions of branched chain amino acids: a review. J Anim Sci Biotechnol 2017; 8:10. [PMID: 28127425 PMCID: PMC5260006 DOI: 10.1186/s40104-016-0139-z] [Citation(s) in RCA: 343] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/27/2016] [Indexed: 02/07/2023] Open
Abstract
It is widely known that branched chain amino acids (BCAA) are not only elementary components for building muscle tissue but also participate in increasing protein synthesis in animals and humans. BCAA (isoleucine, leucine and valine) regulate many key signaling pathways, the most classic of which is the activation of the mTOR signaling pathway. This signaling pathway connects many diverse physiological and metabolic roles. Recent years have witnessed many striking developments in determining the novel functions of BCAA including: (1) Insufficient or excessive levels of BCAA in the diet enhances lipolysis. (2) BCAA, especially isoleucine, play a major role in enhancing glucose consumption and utilization by up-regulating intestinal and muscular glucose transporters. (3) Supplementation of leucine in the diet enhances meat quality in finishing pigs. (4) BCAA are beneficial for mammary health, milk quality and embryo growth. (5) BCAA enhance intestinal development, intestinal amino acid transportation and mucin production. (6) BCAA participate in up-regulating innate and adaptive immune responses. In addition, abnormally elevated BCAA levels in the blood (decreased BCAA catabolism) are a good biomarker for the early detection of obesity, diabetes and other metabolic diseases. This review will provide some insights into these novel metabolic and physiological functions of BCAA.
Collapse
Affiliation(s)
- Shihai Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No.2 Yuanmingyuan West Road, Haidian District, Beijing, 100193 People's Republic of China.,College of Animal Science, South China Agricultural University, Wushan Avenue, Tianhe District, Guangzhou, 510642 People's Republic of China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No.2 Yuanmingyuan West Road, Haidian District, Beijing, 100193 People's Republic of China
| | - Man Ren
- College of Animal Science, Anhui Science & Technology University, No. 9 Donghua Road, Fengyang, 233100 Anhui Province People's Republic of China
| | - Xiangbing Mao
- Animal Nutrition Institute, Key Laboratory of Animal Disease-ResistanceNutrition,Ministry of Education, Sichuan AgriculturalUniversity, Ya'an, Sichuan China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No.2 Yuanmingyuan West Road, Haidian District, Beijing, 100193 People's Republic of China
| |
Collapse
|
22
|
Takami T, Yamasaki T, Saeki I, Matsumoto T, Suehiro Y, Sakaida I. Supportive therapies for prevention of hepatocellular carcinoma recurrence and preservation of liver function. World J Gastroenterol 2016; 22:7252-7263. [PMID: 27621572 PMCID: PMC4997645 DOI: 10.3748/wjg.v22.i32.7252] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/07/2016] [Accepted: 07/21/2016] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest cancers in the world and is associated with a high risk of recurrence. The development of a wide range of new therapies is therefore essential. In this study, from the perspective of supportive therapy for the prevention of HCC recurrence and preservation of liver function in HCC patients, we surveyed a variety of different therapeutic agents. We show that branched chain amino acids (BCAA) supplementation and late evening snack with BCAA, strategies that address issues of protein-energy malnutrition, are important for liver cirrhotic patients with HCC. For chemoprevention of HCC recurrence, we show that viral control after radical treatment is important. We also reviewed the therapeutic potential of antiviral drugs, sorafenib, peretinoin, iron chelators. Sorafenib is a kinase inhibitor and a standard therapy in the treatment of advanced HCC. Peretinoin is a vitamin A-like molecule that targets the retinoid nuclear receptor to induce apoptosis and inhibit tumor growth in HCC cells. Iron chelators, such as deferoxamine and deferasirox, act to prevent cancer cell growth. These chelators may have potential as combination therapies in conjunction with peretinoin. Finally, we review the potential inhibitory effect of bone marrow cells on hepatocarcinogenesis.
Collapse
|
23
|
Rittig N, Bach E, Thomsen HH, Johannsen M, Jørgensen JO, Richelsen B, Jessen N, Møller N. Reply: Letter to the editor - A dietary amino acid load causes a transient decrease in the function of human neutrophil granulocytes. Clin Nutr 2016; 35:771. [PMID: 26887700 DOI: 10.1016/j.clnu.2016.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 01/28/2016] [Indexed: 02/07/2023]
Affiliation(s)
- N Rittig
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus C, Denmark.
| | - E Bach
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus C, Denmark
| | - H H Thomsen
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus C, Denmark
| | - M Johannsen
- Section for Forensic Chemistry, Department of Forensic Medicine, Aarhus University Hospital, Brendstrupgaardsvej 100, 8200, Aarhus N, Denmark
| | - J O Jørgensen
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus C, Denmark
| | - B Richelsen
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus C, Denmark
| | - N Jessen
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus C, Denmark; Department of Biochemical Pathology, Institute for Clinical Medicine, Aarhus University Hospital, Denmark
| | - N Møller
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus C, Denmark
| |
Collapse
|