1
|
Quan T, Li R, Gao T. The Intestinal Macrophage-Intestinal Stem Cell Axis in Inflammatory Bowel Diseases: From Pathogenesis to Therapy. Int J Mol Sci 2025; 26:2855. [PMID: 40243444 PMCID: PMC11988290 DOI: 10.3390/ijms26072855] [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: 02/19/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
The gut plays a crucial role in digestion and immunity, so its balance is essential to overall health. This balance relies on dynamic interactions between intestinal epithelial cells, immune cells, and crypt stem cells. Inflammatory bowel disease (IBD), which consists of ulcerative colitis and Crohn's disease, is a chronic relapsing inflammatory disease of the gastrointestinal tract closely related to immune dysfunction. Stem cells, known for their ability to self-renew and differentiate, play an important role in repairing damaged intestinal epithelium and maintaining homeostasis in vivo. Macrophages are key gatekeepers of intestinal immune homeostasis and have a significant impact on IBD. Current research has focused on the link between epithelial cells and stem cells, but interactions with macrophages, which have been recognized as attractive targets for the development of new therapeutic approaches to disease, have been less explored. Recently, the developing field of immunometabolism has reinforced that metabolic reprogramming is a key determinant of macrophage function and subsequent disease progression. The aim of this review is to explore the role of the macrophage-stem cell axis in the maintenance of intestinal homeostasis and to summarize potential approaches to treating IBD by manipulating the cellular metabolism of macrophages, as well as the main opportunities and challenges faced. In summary, our overview provides a framework for understanding the critical role of macrophage immunometabolism in maintaining gut health and potential therapeutic targets.
Collapse
Affiliation(s)
| | | | - Ting Gao
- College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (T.Q.); (R.L.)
| |
Collapse
|
2
|
Li Y, Tang L, Zhao M, Tang R, Fang K, Ge W, Du W. Study on the active components and mechanism of Atractylodis Macrocephalae Rhizoma for invigorating the spleen and tonifying qi based on spectrum-effect relationship and network pharmacology. Biomed Chromatogr 2024; 38:e5870. [PMID: 38664069 DOI: 10.1002/bmc.5870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/05/2024] [Accepted: 03/11/2024] [Indexed: 06/20/2024]
Abstract
Spleen deficiency can lead to various abnormal physiological functions of the spleen. Atractylodis Macrocephalae Rhizoma (AMR) is a traditional Chinese medicine used to invigorate the spleen and tonify qi. The study aimed to identify the primary active components influencing the efficacy of AMR in strengthening the spleen and replenishing qi through spectrum-effect relationship and chemometrics. Network pharmacology was used to investigate the mechanism by which AMR strengthens the spleen and replenishes qi, with molecular docking utilized for validation purposes. The findings indicated that bran-fried AMR exhibited superior efficacy, with atractylenolides and atractylone identified as the primary active constituents. Atractylenolide II emerged as the most influential component impacting the effectiveness of AMR, while the key target was androgen receptor. Furthermore, crucial pathways implicated included the mitogen-activated protein cascade (MAPK) cascade, RNA polymerase II transcription factor activity, ligand-activated sequence-specific DNA binding, and RNA polymerase II sequence-specific DNA-binding transcription factor binding. In summary, our study has identified the primary active components associated with the efficacy of AMR and has provided an initial exploration of its mechanism of action. This offers a theoretical foundation for future investigations into the material basis and molecular mechanisms underlying the pharmacodynamics of AMR.
Collapse
Affiliation(s)
- Yafei Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Research Center of TCM Processing Technology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lulu Tang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Research Center of TCM Processing Technology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mingfang Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Research Center of TCM Processing Technology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rui Tang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Research Center of TCM Processing Technology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Keer Fang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Research Center of TCM Processing Technology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weihong Ge
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Research Center of TCM Processing Technology, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University Chinese Medicine Yinpian Co., Ltd., Hangzhou, China
| | - Weifeng Du
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Research Center of TCM Processing Technology, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University Chinese Medicine Yinpian Co., Ltd., Hangzhou, China
| |
Collapse
|
3
|
Fu Q, Jiang J, Li X, Zhai Z, Wang X, Li C, Chen Q, Man C, Du L, Wang F, Chen S. Activation of MyD88-Dependent TLR Signaling Modulates Immune Response of the Mouse Heart during Pasteurella multocida Infection. Microorganisms 2023; 11:microorganisms11020400. [PMID: 36838365 PMCID: PMC9967429 DOI: 10.3390/microorganisms11020400] [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: 01/06/2023] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Pasteurella multocida (P. multocida) is an important zoonotic pathogen. In addition to lung lesions, necropsies have revealed macroscopic lesions in the heart in clinical cases. However, most previous studies focused on lung lesions while ignoring heart lesions. Therefore, to investigate the immune response of the P. multocida-infected heart, two murine infection models were established by using P. multocida serotype A (Pm HN02) and D (Pm HN01) strains. Histopathological examination revealed heterogeneous inflammatory responses, including immune cell infiltration in the epicardial and myocardial areas of the heart. Transcriptome sequencing was performed on infected cardiac tissues. To explore the traits of immune responses, we performed the functional enrichment analysis of differentially expressed genes, gene set enrichment analysis and gene set variation analysis. The results showed that the innate immune pathways were significantly regulated in both groups, including the NOD-like receptor signaling pathway, the complement and coagulation cascade and cytokine-cytokine receptor interaction. The Toll-like receptor signaling pathway was only significantly activated in the Pm HN02 group. For the Pm HN02 group, immunohistochemistry analysis further verified the significant upregulation of the hub component MyD88 at the protein level. In conclusion, this study reveals critical pathways for host heart recognition and defense against P. multocida serotypes A and D. Moreover, MyD88 was upregulated by P. multocida serotype A in the heart, providing a theoretical basis for future prevention, diagnosis and treatment research.
Collapse
|
4
|
Macrophage immunometabolism in inflammatory bowel diseases: From pathogenesis to therapy. Pharmacol Ther 2022; 238:108176. [DOI: 10.1016/j.pharmthera.2022.108176] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/11/2022] [Accepted: 03/22/2022] [Indexed: 12/17/2022]
|
5
|
|
6
|
The Amino Acid-mTORC1 Pathway Mediates APEC TW-XM-Induced Inflammation in bEnd.3 Cells. Int J Mol Sci 2021; 22:ijms22179245. [PMID: 34502151 PMCID: PMC8431488 DOI: 10.3390/ijms22179245] [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: 07/13/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
The blood-brain barrier (BBB) is key to establishing and maintaining homeostasis in the central nervous system (CNS); meningitis bacterial infection can disrupt the integrity of BBB by inducing an inflammatory response. The changes in the cerebral uptake of amino acids may contribute to inflammatory response during infection and were accompanied by high expression of amino acid transporters leading to increased amino acid uptake. However, it is unclear whether amino acid uptake is changed and how to affect inflammatory responses in mouse brain microvascular endothelial (bEnd.3) cells in response to Avian Pathogenic Escherichia coli TW-XM (APEC XM) infection. Here, we firstly found that APEC XM infection could induce serine (Ser) and glutamate (Glu) transport from extracellular into intracellular in bEnd.3 cells. Meanwhile, we also shown that the expression sodium-dependent neutral amino acid transporter 2 (SNAT2) for Ser and excitatory amino acid transporter 4 (EAAT4) for Glu was also significantly elevated during infection. Then, in amino acid deficiency or supplementation medium, we found that Ser or Glu transport were involving in increasing SNAT2 or EAAT4 expression, mTORC1 (mechanistic target of rapamycin complex 1) activation and inflammation, respectively. Of note, Ser or Glu transport were inhibited after SNAT2 silencing or EAAT4 silencing, resulting in inhibition of mTORC1 pathway activation, and inflammation compared with the APEC XM infection group. Moreover, pEGFP-SNAT2 overexpression and pEGFP-EAAT4 overexpression in bEnd.3 cells all could promote amino acid uptake, activation of the mTORC1 pathway and inflammation during infection. We further found mTORC1 silencing could inhibit inflammation, the expression of SNAT2 and EAAT4, and amino acid uptake. Taken together, our results demonstrated that APEC TW-XM infection can induce Ser or Glu uptake depending on amino acid transporters transportation, and then activate amino acid-mTORC1 pathway to induce inflammation in bEnd.3 cells.
Collapse
|
7
|
Zhao G, Li P, Mu H, Li N, Peng Y. L-Ascorbic Acid Shapes Bovine Pasteurella multocida Serogroup A Infection. Front Vet Sci 2021; 8:687922. [PMID: 34307527 PMCID: PMC8295749 DOI: 10.3389/fvets.2021.687922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/31/2021] [Indexed: 12/20/2022] Open
Abstract
Bovine Pasteurella multocida serogroup A (bovine PmA) is one of the most important pathogens causing fatal pneumonia in cattle. However, it is largely unknown how nutrition shapes bovine PmA infection. Here, we discovered that the infected lung held the highest bacterial density than other tissues during infection. By screening the different metabolites between high (lung)- and low (liver)-bacterial density tissues, the present work revealed that L-ascorbic acid and L-aspartic acid directly influenced bovine P. multocida growth. Interestingly, L-ascorbic acid, which is expressed at higher levels in the infected livers, inhibited bovine PmA growth as well as virulence factor expression and promoted macrophage bactericidal activity in vitro. In addition, ascorbic acid synthesis was repressed upon bovine PmA infection, and supplementation with exogenous L-ascorbic acid significantly reduced the bacterial burden of the infected lungs and mouse mortality. Collectively, our study has profiled the metabolite difference of the murine lung and liver during bovine PmA infection. The screened L-ascorbic acid showed repression of bovine PmA growth and virulence expression in vitro and supplementation could significantly increase the survival rate of mice and reduce the bacterial load in vivo, which implied that L-ascorbic acid could serve as a potential protective agent for bovine PmA infection in clinic.
Collapse
Affiliation(s)
- Guangfu Zhao
- Chongqing Key Laboratory of Forage and Herbivorce, College of Veterinary Medicine, Southwest University, Chongqing, China
- Key Laboratory for Bio-Resource and Eco-Environment of Education of Ministry, The Center for Growth, Metabolism and Aging, College of Life Sciences, Sichuan University, Chengdu, China
| | - Pan Li
- Chongqing Key Laboratory of Forage and Herbivorce, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Hao Mu
- Chongqing Academy of Animal Science, Chongqing, China
| | - Nengzhang Li
- Chongqing Key Laboratory of Forage and Herbivorce, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yuanyi Peng
- Chongqing Key Laboratory of Forage and Herbivorce, College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
8
|
Beale DJ, Shah R, Karpe AV, Hillyer KE, McAuley AJ, Au GG, Marsh GA, Vasan SS. Metabolic Profiling from an Asymptomatic Ferret Model of SARS-CoV-2 Infection. Metabolites 2021; 11:327. [PMID: 34069591 PMCID: PMC8160988 DOI: 10.3390/metabo11050327] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/06/2021] [Accepted: 05/15/2021] [Indexed: 12/16/2022] Open
Abstract
Coronavirus disease (COVID-19) is a contagious respiratory disease that is causing significant global morbidity and mortality. Understanding the impact of the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection on the host metabolism is still in its infancy but of great importance. Herein, we investigated the metabolic response during viral shedding and post-shedding in an asymptomatic SARS-CoV-2 ferret model (n = 6) challenged with two SARS-CoV-2 isolates. Virological and metabolic analyses were performed on (minimally invasive) collected oral swabs, rectal swabs, and nasal washes. Fragments of SARS-CoV-2 RNA were only found in the nasal wash samples in four of the six ferrets, and in the samples collected 3 to 9 days post-infection (referred to as viral shedding). Central carbon metabolism metabolites were analyzed during viral shedding and post-shedding periods using a dynamic Multiple Reaction Monitoring (dMRM) database and method. Subsequent untargeted metabolomics and lipidomics of the same samples were performed using a Liquid Chromatography Quadrupole Time-of-Flight Mass Spectrometry (LC-QToF-MS) methodology, building upon the identified differentiated central carbon metabolism metabolites. Multivariate analysis of the acquired data identified 29 significant metabolites and three lipids that were subjected to pathway enrichment and impact analysis. The presence of viral shedding coincided with the challenge dose administered and significant changes in the citric acid cycle, purine metabolism, and pentose phosphate pathways, amongst others, in the host nasal wash samples. An elevated immune response in the host was also observed between the two isolates studied. These results support other metabolomic-based findings in clinical observational studies and indicate the utility of metabolomics applied to ferrets for further COVID-19 research that advances early diagnosis of asymptomatic and mild clinical COVID-19 infections, in addition to assessing the effectiveness of new or repurposed drug therapies.
Collapse
Affiliation(s)
- David J. Beale
- Land & Water, Commonwealth Scientific and Industrial Research Organisation, Dutton Park, QLD 4102, Australia or (R.S.); (A.V.K.); (K.E.H.)
| | - Rohan Shah
- Land & Water, Commonwealth Scientific and Industrial Research Organisation, Dutton Park, QLD 4102, Australia or (R.S.); (A.V.K.); (K.E.H.)
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Avinash V. Karpe
- Land & Water, Commonwealth Scientific and Industrial Research Organisation, Dutton Park, QLD 4102, Australia or (R.S.); (A.V.K.); (K.E.H.)
| | - Katie E. Hillyer
- Land & Water, Commonwealth Scientific and Industrial Research Organisation, Dutton Park, QLD 4102, Australia or (R.S.); (A.V.K.); (K.E.H.)
| | - Alexander J. McAuley
- Australian Centre for Disease Preparedness (ACDP), Commonwealth Scientific and Industrial Research Organisation, Geelong, VIC 3220, Australia; (A.J.M.); (G.G.A.); (G.A.M.); (S.S.V.)
| | - Gough G. Au
- Australian Centre for Disease Preparedness (ACDP), Commonwealth Scientific and Industrial Research Organisation, Geelong, VIC 3220, Australia; (A.J.M.); (G.G.A.); (G.A.M.); (S.S.V.)
| | - Glenn A. Marsh
- Australian Centre for Disease Preparedness (ACDP), Commonwealth Scientific and Industrial Research Organisation, Geelong, VIC 3220, Australia; (A.J.M.); (G.G.A.); (G.A.M.); (S.S.V.)
| | - Seshadri S. Vasan
- Australian Centre for Disease Preparedness (ACDP), Commonwealth Scientific and Industrial Research Organisation, Geelong, VIC 3220, Australia; (A.J.M.); (G.G.A.); (G.A.M.); (S.S.V.)
- Department of Health Sciences, University of York, York YO10 5DD, UK
| |
Collapse
|
9
|
Paixão V, Almeida EB, Amaral JB, Roseira T, Monteiro FR, Foster R, Sperandio A, Rossi M, Amirato GR, Santos CAF, Pires RS, Leal FB, Durigon EL, Oliveira DBL, Vieira RP, Vaisberg M, Santos JMB, Bachi ALL. Elderly Subjects Supplemented with L-Glutamine Shows an Improvement of Mucosal Immunity in the Upper Airways in Response to Influenza Virus Vaccination. Vaccines (Basel) 2021; 9:107. [PMID: 33572639 PMCID: PMC7911866 DOI: 10.3390/vaccines9020107] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Although glutamine is able to improve the immune response, its action in the upper airway immunity against the influenza virus vaccine remains unclear. Therefore, we aimed to evaluate the L-glutamine supplementation effect on the mucosal immune/inflammatory response of elderly subjects vaccinated against the influenza virus. METHODS Saliva sampling from 83 physically active elderly volunteers were collected pre- and 30 days after influenza virus vaccination and supplementation with L-glutamine (Gln, n = 42) or placebo (PL, n = 41). RESULTS Gln group showed higher salivary levels of interleukin (IL)-17, total secretory immunoglobulin A (SIgA), and specific-SIgA post-vaccination than values found pre-vaccination and in the PL group post-vaccination. Whereas higher salivary levels of IL-6 and IL-10 were observed post-vaccination in the Gln group, IL-37 levels were lower post-vaccination in both groups than the values pre-vaccination. Tumor necrosis factor (TNF)-α levels were unchanged. Positive correlations between IL-6 and IL-10 were found in all volunteer groups pre- and post-vaccination and also between IL-17 and IL-6 or IL-10 in the Gln group post-vaccination. A negative correlation between IL-37 and IL-10 was found pre- and post-vaccination in the PL group. CONCLUSION Gln supplementation was able to modulate salivary cytokine profile and increase SIgA levels, both total and specific to the influenza virus vaccine, in physically active elderly subjects.
Collapse
Affiliation(s)
- Vitória Paixão
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil; (V.P.); (E.B.A.); (J.B.A.); (T.R.); (F.R.M.); (R.F.); (M.R.); (G.R.A.); (M.V.); (A.L.L.B.)
| | - Ewin B. Almeida
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil; (V.P.); (E.B.A.); (J.B.A.); (T.R.); (F.R.M.); (R.F.); (M.R.); (G.R.A.); (M.V.); (A.L.L.B.)
| | - Jonatas B. Amaral
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil; (V.P.); (E.B.A.); (J.B.A.); (T.R.); (F.R.M.); (R.F.); (M.R.); (G.R.A.); (M.V.); (A.L.L.B.)
| | - Tamaris Roseira
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil; (V.P.); (E.B.A.); (J.B.A.); (T.R.); (F.R.M.); (R.F.); (M.R.); (G.R.A.); (M.V.); (A.L.L.B.)
- Method Faculty of São Paulo (FAMESP), São Paulo 04046-200, Brazil;
| | - Fernanda R. Monteiro
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil; (V.P.); (E.B.A.); (J.B.A.); (T.R.); (F.R.M.); (R.F.); (M.R.); (G.R.A.); (M.V.); (A.L.L.B.)
- Method Faculty of São Paulo (FAMESP), São Paulo 04046-200, Brazil;
| | - Roberta Foster
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil; (V.P.); (E.B.A.); (J.B.A.); (T.R.); (F.R.M.); (R.F.); (M.R.); (G.R.A.); (M.V.); (A.L.L.B.)
- Method Faculty of São Paulo (FAMESP), São Paulo 04046-200, Brazil;
| | | | - Marcelo Rossi
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil; (V.P.); (E.B.A.); (J.B.A.); (T.R.); (F.R.M.); (R.F.); (M.R.); (G.R.A.); (M.V.); (A.L.L.B.)
| | - Gislene R. Amirato
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil; (V.P.); (E.B.A.); (J.B.A.); (T.R.); (F.R.M.); (R.F.); (M.R.); (G.R.A.); (M.V.); (A.L.L.B.)
| | - Carlos A. F. Santos
- Department of Medicine, Geriatry, Paulista School of Medicine (EPM), São Paulo 04023-062, Brazil;
| | - Renier S. Pires
- Post-Graduation Program in Health Science, Santo Amaro University (UNISA), São Paulo 04743-030, Brazil;
| | - Fabyano B. Leal
- Institute of Biomedical Science, University of São Paulo (USP), São Paulo 05508-060, Brazil; (F.B.L.); (E.L.D.); (D.B.L.O.)
| | - Edison L. Durigon
- Institute of Biomedical Science, University of São Paulo (USP), São Paulo 05508-060, Brazil; (F.B.L.); (E.L.D.); (D.B.L.O.)
- Scientific Platform Pasteur, University of São Paulo (USP), São Paulo 05508-060, Brazil
| | - Danielle B. L. Oliveira
- Institute of Biomedical Science, University of São Paulo (USP), São Paulo 05508-060, Brazil; (F.B.L.); (E.L.D.); (D.B.L.O.)
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil
| | - Rodolfo P. Vieira
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), São Paulo 12245-520, Brazil;
- Post-Graduation Program in Bioengineering and Biomedical Engineering, Universidade Brasil, São Paulo 15600-000, Brazil
- Post-Graduation Program in Science of Human and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos 11015-020, Brazil
| | - Mauro Vaisberg
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil; (V.P.); (E.B.A.); (J.B.A.); (T.R.); (F.R.M.); (R.F.); (M.R.); (G.R.A.); (M.V.); (A.L.L.B.)
| | - Juliana M. B. Santos
- Post-Graduation Program in Science of Human and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos 11015-020, Brazil
| | - André L. L. Bachi
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil; (V.P.); (E.B.A.); (J.B.A.); (T.R.); (F.R.M.); (R.F.); (M.R.); (G.R.A.); (M.V.); (A.L.L.B.)
- Post-Graduation Program in Health Science, Santo Amaro University (UNISA), São Paulo 04743-030, Brazil;
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), São Paulo 12245-520, Brazil;
| |
Collapse
|
10
|
Yuan D, Wang J, Xiao D, Li J, Liu Y, Tan B, Yin Y. Eucommia ulmoides Flavones as Potential Alternatives to Antibiotic Growth Promoters in a Low-Protein Diet Improve Growth Performance and Intestinal Health in Weaning Piglets. Animals (Basel) 2020; 10:E1998. [PMID: 33143126 PMCID: PMC7694009 DOI: 10.3390/ani10111998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 02/08/2023] Open
Abstract
Eucommia ulmoides flavones (EUF) have been demonstrated to attenuate the inflammation and oxidative stress of piglets. This study aimed to test whether EUF could be used as an alternative antibiotic growth promoter to support growth performance and maintain intestinal health in weanling piglets. Weaned piglets (n = 480) were assigned into three groups and fed with a low-protein basal diet (NC), or supplementation with antibiotics (PC) or 0.01% EUF (EUF). Blood, intestinal contents, and intestine were collected on days 15 and 35 after weaning. The results showed the PC and EUF supplementations increased (p < 0.05) body weight on day 35, average daily gain and gain: feed ratio from day 15 to day 35 and day 0 to day 35, whereas decreased (p < 0.05) the diarrhea index of weanling piglets. EUF treatment increased (p < 0.05) jejunal villus height: crypt depth ratio, jejunal and ileal villus height, and population of ileal lactic acid bacteria on day 15 but decreased (p < 0.05) the population of ileal coliform bacteria on day 15 and day 35. These findings indicated the EUF, as the potential alternative to in-feed antibiotic growth promoter, could improve growth performance and intestinal morphology, and decrease colonization of coliform bacteria and diarrhea index in weanling piglets.
Collapse
Affiliation(s)
- Daixiu Yuan
- Department of Medicine, Jishou University, Jishou 416000, China;
| | - Jing Wang
- Department of Animal Science, Hunan Agricultural University, Changsha 410000, China; (D.X.); (B.T.)
- Laboratory of Animal Nutritional Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| | - Dingfu Xiao
- Department of Animal Science, Hunan Agricultural University, Changsha 410000, China; (D.X.); (B.T.)
| | - Jiefeng Li
- Laboratory of Animal Nutritional Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA 95616, USA;
| | - Bie Tan
- Department of Animal Science, Hunan Agricultural University, Changsha 410000, China; (D.X.); (B.T.)
- Laboratory of Animal Nutritional Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| |
Collapse
|
11
|
Xin J, Zeng D, Wang H, Sun N, Khalique A, Zhao Y, Wu L, Pan K, Jing B, Ni X. Lactobacillus johnsonii BS15 improves intestinal environment against fluoride-induced memory impairment in mice-a study based on the gut-brain axis hypothesis. PeerJ 2020; 8:e10125. [PMID: 33083147 PMCID: PMC7547597 DOI: 10.7717/peerj.10125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 09/17/2020] [Indexed: 01/09/2023] Open
Abstract
Background Excessive fluoride can lead to chronic neurodegeneration characterized by neuron and myelin loss and memory dysfunction. The gut–brain axis hypothesis suggests that gut microbiota plays a crucial role in regulating brain function. Thus, using probiotics to adjust the gut microenvironment may be a potential therapy for mental diseases. Methods Mice in the prob group were administrated with Lactobacillus johnsonii BS15 for 28 days prior to and throughout a 70-day exposure to sodium fluoride. The drinking water of all groups (F and prob groups) except the control group were replaced by high-fluoride water (100 mg NaF/L) on day 28. Animals in each group were divided into two subsets: one underwent behavioral test, and the other was sacrificed for sampling. The mRNA expression level and protein content related to inflammatory reaction in the ileum and hippocampus were respectively detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). The mRNA expression levels of proteins related to myelin structure, apoptosis, and memory in the hippocampus and tight junction proteins in the ileum were determined by RT-qPCR and/or immunohistochemistry. Gut permeability markers (D-lactate and diamine oxidase (DAO)) in the serum were also examined by ELISA. Results The results showed that fluoride exposure induced a lower spontaneous exploration (P < 0.05) in T-maze test, which indicated an impairment of memory. Spontaneous exploration of BS15-treated mice was significantly higher (P < 0.05) than that in F group. Fluoride reduced (P < 0.05) levels of myelin structural protein (proteolipid protein) and neurogenesis-associated proteins (brain-derived neurotrophic factor and cAMP/Ca2+ responsive element-binding protein), induced disordered inflammatory cytokines (TNF-α, IFN-γ, and IL-6; P < 0.05), increased pro-apoptotic genes (caspase-3; P < 0.05), and decreased anti-apoptotic genes (Bcl-2; P < 0.05) in the hippocampus, of which the influences were reversed by BS15. BS15 treatment exerted significant preventive effects on reversing the gut inflammation induced by excessive fluoride intake by reducing (P < 0.05) the levels of pro-inflammatory cytokines (tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ)) and remarkably increasing (P < 0.05) the level of anti-inflammatory cytokines (IL-10). Moreover, the serum DAO activity and D-lactate concentration significantly increased by fluoride were also reduced (P < 0.05) by BS15. This result indicated the profitable effect of BS15 on gut permeability. Conclusion L. johnsonii BS15 intake could benefit the neuroinflammation and demyelination in the hippocampus by improving the gut environment and ameliorating fluorine-induced memory dysfunction.
Collapse
Affiliation(s)
- Jinge Xin
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hesong Wang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Abdul Khalique
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ying Zhao
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Liqian Wu
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Ren W, Bin P, Yin Y, Wu G. Impacts of Amino Acids on the Intestinal Defensive System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1265:133-151. [PMID: 32761574 DOI: 10.1007/978-3-030-45328-2_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The intestine interacts with a diverse community of antigens and bacteria. To keep its homeostasis, the gut has evolved with a complex defense system, including intestinal microbiota, epithelial layer and lamina propria. Various factors (e.g., nutrients) affect the intestinal defensive system and progression of intestinal diseases. This review highlights the current understanding about the role of amino acids (AAs) in protecting the intestine from harm. Amino acids (e.g., arginine, glutamine and tryptophan) are essential for the function of intestinal microbiota, epithelial cells, tight junction, goblet cells, Paneth cells and immune cells (e.g., macrophages, B cells and T cells). Through the modulation of the intestinal defensive system, AAs maintain the integrity and function of the intestinal mucosa and inhibit the progression of various intestinal diseases (e.g., intestinal infection and intestinal colitis). Thus, adequate intake of functional AAs is crucial for intestinal and whole-body health in humans and other animals.
Collapse
Affiliation(s)
- Wenkai Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Peng Bin
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product, Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yulong Yin
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
13
|
J. Ayon N. Features, roles and chiral analyses of proteinogenic amino acids. AIMS MOLECULAR SCIENCE 2020. [DOI: 10.3934/molsci.2020011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
|
14
|
Wang K, Li Y, Dai Y, Han L, Zhu Y, Xue C, Wang P, Wang J. Peptides from Antarctic Krill ( Euphausia superba) Improve Osteoarthritis via Inhibiting HIF-2α-Mediated Death Receptor Apoptosis and Metabolism Regulation in Osteoarthritic Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:3125-3133. [PMID: 30798606 DOI: 10.1021/acs.jafc.8b05841] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Osteoarthritis (OA) is a prevalent debilitating disease which is predominantly characterized by cartilage degeneration. In the current study, destabilization of the medial meniscus (DMM) mouse model was used to investigate the effects of Antarctic krill peptides (AKP) on cartilage protection. As observed, AKP clearly ameliorate cartilage degeneration as evidenced by increased cartilage thickness and cartilage area and decreased histological Osteoarthritis Research Society International (OARSI) scores. Toluidine blue staining showed that AKO remarkably inhibited the loss of cartilage matrix in mice with OA. Hypoxia-inducible factor-2α (HIF-2α) has a key role in catabolic regulation and inflammation cascades which are the main causes of OA. AKP can down-regulate the expression of HIF-2α and its downstream genes such as MMP-13, Adamts-5, IL-1β, iNOS, CXCL-1, and NOS2. Consistent with this, anabolic genes such as Acan and Col2α1 were restored after treatment with AKP. Chondrocyte apoptosis and the reduction in cartilage cell viability are also involved in the process of OA. The HIF-2α-mediated death receptor apoptosis signaling pathway has been involved in the regulation of chondrocyte apoptosis. AKP can reduce the expressions of key pro-apoptosis genes in Fas-FasL and DR3-DR3L signaling pathways such as Fas, FasL, FADD, caspase8, caspase3, DR3, DR3L, RIP, and NF-κB. In addition, expressions of antiapoptosis genes such as c-AIP and c-FLIP were increased significantly. These findings indicate that AKP can be used as a new functional factor in the development of functional foods and chondroprotective drugs.
Collapse
Affiliation(s)
- Kai Wang
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Yuanyuan Li
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Yufeng Dai
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Lihau Han
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Yujie Zhu
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Changhu Xue
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Peng Wang
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Jingfeng Wang
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| |
Collapse
|
15
|
Yang B, Ni J, Long H, Huang J, Yang C, Huang X. IL-1β-induced miR-34a up-regulation inhibits Cyr61 to modulate osteoarthritis chondrocyte proliferation through ADAMTS-4. J Cell Biochem 2018; 119:7959-7970. [PMID: 29236314 DOI: 10.1002/jcb.26600] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/04/2017] [Indexed: 01/13/2023]
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease with multifactorial etiology caused by risk factors. The degradation of aggrecan by upregulated ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) is the key event in the development of OA. ADAMTS-4 contributes to aggrecan degradation in human OA. Cysteine-rich angiogenic inducer 61 (Cyr61), which is associated with diseases related to chronic inflammation, is found in articular cartilage from patients with osteoarthritis and appears to suppress ADAMTS-4 activity, possibly leading to chondrocyte cloning. Herein, we first revealed that Cyr61 and ADAMTS-4 protein levels were remarkably increased in OA cartilage tissues and OA chondrocytes, and verified Cyr61 regulation of ADAMTS-4 in normal and OA chondrocyte. Further, we revealed that Cyr61 could promote OA chondrocyte proliferation through inhibiting ADAMTS-4. Overproduction of inflammatory cytokines plays a vital role in the pathological development of OA; herein, we demonstrated that IL-1β inhibited Cyr61, while promoted ADAMTS-4 expression. By using online tools and luciferase assays, we confirmed that miR-34a, a regulatory miRNA of chondrocyte proliferation, could directly bind to the 3'-UTR of Cyr61 to inhibit its expression; further, IL-1β regulated Cyr61 and ADAMTS-4 expression through miR-34a. In OA cartilage tissues, miR-34a, and IL-1β mRNA expression was up-regulated and positively correlated; miR-34a and Cyr61 mRNA was positively correlated, further indicating that suppressing miR-34a expression might rescue IL-1β-induced Cyr61 suppression, and promote OA chondrocyte proliferation. Taken together, we provided novel experimental basis for rescuing OA chondrocyte proliferation through miR-34a/Cyr61 axis.
Collapse
Affiliation(s)
- Bo Yang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Jiangdong Ni
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Hui Long
- Department of Pain, The Second Affiliated Hospital of Nanhua University, Hengyang, Hunan, P.R. China
| | - Jun Huang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Cheng Yang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xianzhe Huang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
16
|
Chen S, Bin P, Ren W, Gao W, Liu G, Yin J, Duan J, Li Y, Yao K, Huang R, Tan B, Yin Y. Alpha-ketoglutarate (AKG) lowers body weight and affects intestinal innate immunity through influencing intestinal microbiota. Oncotarget 2018; 8:38184-38192. [PMID: 28465471 PMCID: PMC5503525 DOI: 10.18632/oncotarget.17132] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 04/04/2017] [Indexed: 12/17/2022] Open
Abstract
Alpha-ketoglutarate (AKG), a precursor of glutamate and a critical intermediate in the tricarboxylic acid cycle, shows beneficial effects on intestinal function. However, the influence of AKG on the intestinal innate immune system and intestinal microbiota is unknown. This study explores the effect of oral AKG administration in drinking water (10 g/L) on intestinal innate immunity and intestinal microbiota in a mouse model. Mouse water intake, feed intake and body weight were recorded throughout the entire experiment. The ileum was collected for detecting the expression of intestinal proinflammatory cytokines and innate immune factors by Real-time Polymerase Chain Reaction. Additionally, the ileal luminal contents and feces were collected for 16S rDNA sequencing to analyze the microbial composition. The intestinal microbiota in mice was disrupted with an antibiotic cocktail. The results revealed that AKG supplementation lowered body weight, promoted ileal expression of mammalian defensins of the alpha subfamily (such as cryptdins-1, cryptdins-4, and cryptdins-5) while influencing the intestinal microbial composition (i.e., lowering the Firmicutes to Bacteroidetes ratio). In the antibiotic-treated mouse model, AKG supplementation failed to affect mouse body weight and inhibited the expression of cryptdins-1 and cryptdins-5 in the ileum. We concluded that AKG might affect body weight and intestinal innate immunity through influencing intestinal microbiota.
Collapse
Affiliation(s)
- Shuai Chen
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Peng Bin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenkai Ren
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wei Gao
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China
| | - Gang Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China
| | - Jie Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jielin Duan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yinghui Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Kang Yao
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China
| | - Ruilin Huang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China
| | - Bie Tan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Changsha, Hunan, China
| |
Collapse
|
17
|
Autophagy Strengthens Intestinal Mucosal Barrier by Attenuating Oxidative Stress in Severe Acute Pancreatitis. Dig Dis Sci 2018; 63:910-919. [PMID: 29427225 DOI: 10.1007/s10620-018-4962-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/01/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Intestinal mucosal barrier dysfunction can be caused by severe acute pancreatitis (SAP). It is normally associated with changes to mucosal autophagy and oxidative stress. OBJECTIVE The aim of this study was to investigate the correlation between autophagy and oxidative stress on the intestinal mucosal barrier of SAP rat model. METHODS SAP was induced by retrograde injection of sodium taurocholate (5%) into the biliopancreatic duct. Bacterial translocation (BT) was detected by 16S rDNA sequencing analysis. Morphological alterations in the pancreas and gut were determined by hematoxylin-eosin staining. Oxidative stress status was determined by measuring the level of intestinal malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione peroxidase (GPx). Western blot, RT-PCR, and immunofluorescent staining were preformed to analyze the expression of tight junction and autophagy proteins. RESULTS According to the sequencing analysis, rats in SAP group were divided into BT (+) group (n = 9) and BT (-) group (n = 8). Pancreatic and intestinal injuries in SAP group were significantly higher than sham operation group. The content of MDA was clearly elevated, and SOD as well as GPx activities were decreased in BT (+) group as compared with BT (-) group. The expression of LC3II and Beclin1 in BT (-) group was higher than that observed in BT (+). In contrast, BT (+) group had a higher level of claudin-2 and a lower level of zonula occluden-1, occludin, and claudin-1. CONCLUSION These results suggest that activated autophagy may attenuate intestinal mucosal barrier dysfunction by preventing and reducing the oxidative stress in SAP.
Collapse
|
18
|
Ren W, Rajendran R, Zhao Y, Tan B, Wu G, Bazer FW, Zhu G, Peng Y, Huang X, Deng J, Yin Y. Amino Acids As Mediators of Metabolic Cross Talk between Host and Pathogen. Front Immunol 2018. [PMID: 29535717 PMCID: PMC5835074 DOI: 10.3389/fimmu.2018.00319] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The interaction between host and pathogen decidedly shapes the outcome of an infection, thus understanding this interaction is critical to the treatment of a pathogen-induced infection. Although research in this area of cell biology has yielded surprising findings regarding interactions between host and pathogen, understanding of the metabolic cross talk between host and pathogen is limited. At the site of infection, host and pathogen share similar or identical nutritional substrates and generate common metabolic products, thus metabolic cross talk between host and pathogen could profoundly affect the pathogenesis of an infection. In this review, we present results of a recent discovery of a metabolic interaction between host and pathogen from an amino acid (AA) metabolism-centric point of view. The host depends on AA metabolism to support defensive responses against pathogens, while the pathogens modulate AA metabolism for its own advantage. Some AA, such as arginine, asparagine, and tryptophan, are central points of competition between the host and pathogen. Thus, a better understanding of AA-mediated metabolic cross talk between host and pathogen will provide insight into fruitful therapeutic approaches to manipulate and prevent progression of an infection.
Collapse
Affiliation(s)
- Wenkai Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China.,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Ranjith Rajendran
- School of Medicine, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow, United Kingdom
| | - Yuanyuan Zhao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Bie Tan
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, TAMU, College Station, TX, United States
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, TAMU, College Station, TX, United States
| | - Guoqiang Zhu
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yuanyi Peng
- Chongqing Key Laboratory of Forage & Herbivorce, College of Animal Science and Technology, Southwest University, Chongqing, China
| | | | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yulong Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
19
|
Liu Q, Ni X, Wang Q, Peng Z, Niu L, Wang H, Zhou Y, Sun H, Pan K, Jing B, Zeng D. Lactobacillus plantarum BSGP201683 Isolated from Giant Panda Feces Attenuated Inflammation and Improved Gut Microflora in Mice Challenged with Enterotoxigenic Escherichia coli. Front Microbiol 2017; 8:1885. [PMID: 29018435 PMCID: PMC5623042 DOI: 10.3389/fmicb.2017.01885] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/14/2017] [Indexed: 12/28/2022] Open
Abstract
In this work, we searched for an effective probiotic that can help control intestinal infection, particularly enterotoxigenic Escherichia coli K88 (ETEC) invasion, in giant panda (Ailuropoda melanoleuca). As a potential probiotic strain, Lactobacillus plantarum BSGP201683 (L. plantarum G83) was isolated from the feces of giant panda and proven beneficial in vitro. This study was aimed to evaluate the protective effect of L. plantarum G83 in mice challenged with ETEC. The mice were orally administered with 0.2 mL of PBS containing L. plantarum G83 at 0 colony-forming units (cfu) mL−1 (control; negative control, ETEC group), 5.0 × 108 cfu mL−1 (LDLP), 5.0 × 109 cfu mL−1 (MDLP), and 5.0 × 1010 cfu mL−1 (HDLP) for 14 consecutive days. At day 15, the mice (LDLP, MDLP, HDLP, and ETEC groups) were challenged with ETEC and assessed at 0, 24, and 144 h. Animal health status; chemical and biological intestinal barriers; and body weight were measured. Results showed that L. plantarum G83 supplementation protected the mouse gut mainly by attenuating inflammation and improving the gut microflora. Most indices significantly changed at 24 h after challenge compared to those at 0 and 144 h. All treatment groups showed inhibited plasma diamine oxidase activity and D-lactate concentration. Tight-junction protein expression was down-regulated, and interleukin (IL)-1β, IL-6, IL-8, TLR4, and MyD88 levels were up-regulated in the jejunum in the LDLP and MDLP groups. The number of the Enterobacteriaceae family and the heat-labile enterotoxin (LT) gene decreased (P < 0.05) in the colons in the LDLP and MDLP groups. All data indicated that L. plantarum G83 could attenuate acute intestinal inflammation caused by ETEC infection, and the low and intermediate doses were superior to the high dose. These findings suggested that L. plantarum G83 may serve as a protective probiotic for intestinal disease and merits further investigation.
Collapse
Affiliation(s)
- Qian Liu
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qiang Wang
- Chengdu Wildlife Institute, Chengdu Zoo, Chengdu, China
| | - Zhirong Peng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lili Niu
- Chengdu Wildlife Institute, Chengdu Zoo, Chengdu, China
| | - Hengsong Wang
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yi Zhou
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hao Sun
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Kangcheng Pan
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Bo Jing
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
20
|
Rom O, Grajeda-Iglesias C, Najjar M, Abu-Saleh N, Volkova N, Dar DE, Hayek T, Aviram M. Atherogenicity of amino acids in the lipid-laden macrophage model system in vitro and in atherosclerotic mice: a key role for triglyceride metabolism. J Nutr Biochem 2017; 45:24-38. [DOI: 10.1016/j.jnutbio.2017.02.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/27/2017] [Accepted: 02/25/2017] [Indexed: 12/27/2022]
|
21
|
Liang J, Xu L, Zhou F, Liu AM, Ge HX, Chen YY, Tu M. MALAT1/miR-127-5p Regulates Osteopontin (OPN)-Mediated Proliferation of Human Chondrocytes Through PI3K/Akt Pathway. J Cell Biochem 2017; 119:431-439. [PMID: 28590075 DOI: 10.1002/jcb.26200] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/06/2017] [Indexed: 01/10/2023]
Abstract
Osteoarthritis (OA) is characterized by progressive destruction of articular cartilage, resulting in significant disability. Chondrocytes present in various types of cartilage and are responsible for the growth and maintenance of the tissue. Over-proliferation of human chondrocytes may contributes to OA pathological process. Previously, we revealed that miR-127-5p could inhibit the proliferation of human chondrocytes through osteopontin (OPN). In the present study, we used online tools to figure out several candidates lncRNAs which were potentially correlated with miR-127-5p. Through assessing the expression levels of the candidates lncRNAs, metastasis associated lung adenocarcinoma transcript 1 (MALAT1) was chosen as a further research subject. MALAT1 knockdown significantly repressed human OA chondrocyte proliferation, as well as the protein levels of OPN, p-PI3K, and p-Akt in OA chondrocytes. As verified by luciferase assays, MALAT1 directly bound to miR-127-5p to inhibit miR-127-5p expression. Then we achieved miR-127-5p inhibition through miR-127-5p inhibitor transfection; the miR-127-5p inhibition could promote chondrocyte proliferation, as well as the protein levels of OPN, p-PI3K, and p-Akt; in addition, the MALAT1 knockdown partially reversed the promotive effect of miR-127-5p inhibition on chondrocyte proliferation, OPN and PI3K/Akt signaling-related protein levels. Taken together, MALAT1 could directly bind to miR-127-5p to inhibit its expression, so as to rescue OPN expression and promote chondrocyte proliferation through PI3K/Akt pathway. Targeting MALAT1 so as to rescue miR-127-5p expression in OA might help to inhibit chondrocyte proliferation through miR-127-5p-mediated OPN regulation and downstream PI3K/Akt pathway. J. Cell. Biochem. 119: 431-439, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jun Liang
- Department of Orthopedics, Second People's Hospital of Jingmen, Jingmen 448000, China
| | - Li Xu
- Department of Orthopedics, Second People's Hospital of Jingmen, Jingmen 448000, China
| | - Feng Zhou
- Department of Orthopedics, Second People's Hospital of Jingmen, Jingmen 448000, China
| | - An-Min Liu
- Department of Orthopedics, Second People's Hospital of Jingmen, Jingmen 448000, China
| | - Hong-Xing Ge
- Department of Orthopedics, Second People's Hospital of Jingmen, Jingmen 448000, China
| | - Yuan-Yuan Chen
- Department of Orthopedics, Second People's Hospital of Jingmen, Jingmen 448000, China
| | - Min Tu
- Department of Orthopedics, Second People's Hospital of Jingmen, Jingmen 448000, China
| |
Collapse
|
22
|
Wu C, Qin X, Li P, Pan T, Ren W, Li N, Peng Y. Transcriptomic Analysis on Responses of Murine Lungs to Pasteurella multocida Infection. Front Cell Infect Microbiol 2017; 7:251. [PMID: 28676843 PMCID: PMC5476747 DOI: 10.3389/fcimb.2017.00251] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/30/2017] [Indexed: 01/09/2023] Open
Abstract
Pasteurella multocida infection in cattle causes serious epidemic diseases and leads to great economic losses in livestock industry; however, little is known about the interaction between host and P. multocida in the lungs. To explore a fully insight into the host responses in the lungs during P. multocida infection, a mouse model of Pasteurella pneumonia was established by intraperitoneal infection, and then transcriptomic analysis of infected lungs was performed. P. multocida localized and grew in murine lungs, and induced inflammation in the lungs, as well as mice death. With transcriptomic analysis, approximately 107 clean reads were acquired. 4236 differently expressed genes (DEGs) were detected during P. multocida infection, of which 1924 DEGs were up-regulated. By gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichments, 5,303 GO enrichments and 116 KEGG pathways were significantly enriched in the context of P. multocida infection. Interestingly, genes related to immune responses, such as pattern recognition receptors (PRRs), chemokines and inflammatory cytokines, were significantly up-regulated, suggesting the key roles of these genes in P. multocida infection. Transcriptomic data showed that IFN-γ/IL-17-related genes were increased, which were validated by qRT-PCR, ELISA, and immunoblotting. Our study characterized the transcriptomic profile of the lungs in mice upon Pasteurella infection, and our findings could provide valuable information with respect to better understanding the responses in mice during P. multocida infection.
Collapse
Affiliation(s)
- Chenlu Wu
- College of Animal Science and Technology, Southwest UniversityChongqing, China
| | - Xiaobin Qin
- College of Animal Science and Technology, Southwest UniversityChongqing, China
| | - Pan Li
- College of Animal Science and Technology, Southwest UniversityChongqing, China
| | - Tingting Pan
- College of Animal Science and Technology, Southwest UniversityChongqing, China
| | - Wenkai Ren
- Key Laboratory of Agro-ecological Processes in Subtropical Region, 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, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangsha, China
| | - Nengzhang Li
- College of Animal Science and Technology, Southwest UniversityChongqing, China
| | - Yuanyi Peng
- College of Animal Science and Technology, Southwest UniversityChongqing, China
| |
Collapse
|
23
|
Wang Q, Wang W, Zhang F, Deng Y, Long Z. NEAT1/miR-181c Regulates Osteopontin (OPN)-Mediated Synoviocyte Proliferation in Osteoarthritis. J Cell Biochem 2017; 118:3775-3784. [PMID: 28379604 DOI: 10.1002/jcb.26025] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 03/30/2017] [Indexed: 01/21/2023]
Abstract
Osteoarthritis (OA) is characterized by progressive destruction of articular cartilage, resulting in significant disability. Inflammatory cytokines commonly initiate the extreme changes in the synovium and cartilage microenvironment of the OA patients, subsequently resulting in cell dysfunctions, especially synoviocyte dysfunction. We revealed that the expression of osteopontin (OPN), which has been reported to regulate expression of various inflammatory factors associating with the pathogenesis of OA including matrix metalloprotease 13 (MMP13), interlukine-6 and 8 (IL-6 and IL-8), is significantly upregulated in OA tissues. In the present study, online tools were used to screen out the candidate miRNAs of OPN. Among the candidate miRNAs, miR-181c inhibited OPN mRNA expression the most strongly. Ectopic expression of miR-181c significantly repressed synoviocyte proliferation, as well as the levels of OPN, MMP13, IL-6, and IL-8. Further, the candidate lncRNAs of miR-181c were screened out by using DianaTools; among which NEAT1 showed to inversely regulate miR-181c. By performing Luciferase assays, we revealed that NEAT1 competed with OPN for miR-181c binding. After NEAT1 knockdown, MMP13, IL-6, and IL-8 expression was reduced; the synoviocyte proliferation was repressed, as well as OPN protein levels; the suppressive effect of NETA1 knockdown on synoviocyte proliferation and the indicated factors were partially reversed by miR-181c inhibition. In OA tissues, OPN mRNA, and NEAT1 expression was upregulated, whereas miR-181c expression was downregulated, indicating that targeting NEAT1 to rescue miR-181c expression so as to inhibit OPN expression and synoviocyte proliferation might be an efficient strategy for OA treatment. J. Cell. Biochem. 118: 3775-3784, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Qiyuan Wang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, Hunan, China
| | - Wanchun Wang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fan Zhang
- Department of Neonatology, The Hunan Children's Hospital, Changsha, Hunan, China
| | - Youwen Deng
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, Hunan, China
| | - Zeling Long
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Liu G, Ren W, Fang J, Hu CAA, Guan G, Al-Dhabi NA, Yin J, Duraipandiyan V, Chen S, Peng Y, Yin Y. L-Glutamine and L-arginine protect against enterotoxigenic Escherichia coli infection via intestinal innate immunity in mice. Amino Acids 2017; 49:1945-1954. [PMID: 28299479 DOI: 10.1007/s00726-017-2410-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/04/2017] [Indexed: 12/13/2022]
Abstract
Dietary glutamine (Gln) or arginine (Arg) supplementation is beneficial for intestinal health; however, whether Gln or Arg may confer protection against Enterotoxigenic Escherichia coli (ETEC) infection is not known. To address this, we used an ETEC-infected murine model to investigate the protective effects of Gln and Arg. Experimentally, we pre-treated mice with designed diet of Gln or Arg supplementation prior to the oral ETEC infection and then assessed mouse mortality and intestinal bacterial burden. We also determined the markers of intestinal innate immunity in treated mice, including secretory IgA response (SIgA), mucins from goblet cells, as well as antimicrobial peptides from Paneth cells. ETEC colonized in mouse small intestine, including duodenum, jejunum, and ileum, and inhibited the mRNA expression of intestinal immune factors, such as polymeric immunoglobulin receptor (pIgR), cryptdin-related sequence 1C (CRS1C), and Reg3γ. We found that dietary Gln or Arg supplementation decreased bacterial colonization and promoted the activation of innate immunity (e.g., the mRNA expression of pIgR, CRS1C, and Reg3γ) in the intestine of ETEC-infected mice. Our results suggest that dietary arginine or glutamine supplementation may inhibit intestinal ETEC infection through intestinal innate immunity.
Collapse
Affiliation(s)
- Gang Liu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Hunan, 410125, People's Republic of China
| | - Wenkai Ren
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Hunan, 410125, People's Republic of China.
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
| | - Chien-An Andy Hu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, MSC08 4670, Fitz 258, Albuquerque, NM, 87131, USA
| | - Guiping Guan
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
| | - Naif Abdullah Al-Dhabi
- Addiriyah Chair for Environmental Studies, Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Jie Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Hunan, 410125, People's Republic of China
| | - Veeramuthu Duraipandiyan
- Addiriyah Chair for Environmental Studies, Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Shuai Chen
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Hunan, 410125, People's Republic of China
| | - Yuanyi Peng
- College of Animal Science and Technology, Southwest University, Chongqing, 400716, People's Republic of China
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Co-Innovation Center of Animal Production Safety, Hunan, 410125, People's Republic of China.
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha, Hunan, People's Republic of China.
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
| |
Collapse
|
25
|
Ren W, Wang K, Yin J, Chen S, Liu G, Tan B, Wu G, Bazer FW, Peng Y, Yin Y. Glutamine-Induced Secretion of Intestinal Secretory Immunoglobulin A: A Mechanistic Perspective. Front Immunol 2016; 7:503. [PMID: 27933057 PMCID: PMC5121228 DOI: 10.3389/fimmu.2016.00503] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/27/2016] [Indexed: 12/23/2022] Open
Abstract
Secretory immunoglobulin A (SIgA) is one important line of defense in the intestinal mucosal surface to protect the intestinal epithelium from enteric toxins and pathogenic microorganisms. Multiple factors, such as intestinal microbiota, intestinal cytokines, and nutrients are highly involved in production of SIgA in the intestine. Recently, glutamine has been shown to affect intestinal SIgA production; however, the underlying mechanism by which glutamine stimulates secretion of intestinal SIgA is unknown. Here, we review current knowledge regarding glutamine in intestinal immunity and show that glutamine-enhanced secretion of SIgA in the intestine may involve intestinal microbiota, intestinal antigen sampling and presentation, induction pathways for SIgA production by plasma cells (both T-dependent and T-independent pathway), and even transport of SIgA. Altogether, the glutamine-intestinal SIgA axis has broad therapeutic implications for intestinal SIgA-associated diseases, such as celiac disease, allergies, and inflammatory bowel disease.
Collapse
Affiliation(s)
- Wenkai Ren
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Kai Wang
- Institute of Apicultural Research (IAR), Chinese Academy of Agricultural Sciences (CAAS) , Beijing , China
| | - Jie Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Shuai Chen
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Gang Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences , Changsha , China
| | - Bie Tan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences , Changsha , China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University , College Station, TX , USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University , College Station, TX , USA
| | - Yuanyi Peng
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science and Technology, Southwest University , Chongqing , China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China; College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
26
|
YANG XIANGWU, XIAO ZHIMING, LIU FEN, CHEN SHUAI, TANG WULIANG, ZHANG DECAI, LIU SHAOJUN. Enterotoxigenic Escherichia coli infection alters intestinal immunity in mice. Mol Med Rep 2016; 14:825-30. [DOI: 10.3892/mmr.2016.5302] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/07/2016] [Indexed: 11/06/2022] Open
|
27
|
Tu M, Li Y, Zeng C, Deng Z, Gao S, Xiao W, Luo W, Jiang W, Li L, Lei G. MicroRNA-127-5p regulates osteopontin expression and osteopontin-mediated proliferation of human chondrocytes. Sci Rep 2016; 6:25032. [PMID: 27126955 PMCID: PMC4850404 DOI: 10.1038/srep25032] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/08/2016] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to determine the specific microRNA (miRNA) that regulates expression of osteopontin (OPN) in osteoarthritis (OA). The potential regulatory miRNAs for OPN messenger RNA (mRNA) were predicted by miRNA prediction programs. Among eight potential regulatory miRNAs, miR-220b, miR-513a-3p and miR-548n increased, while miR-181a, miR-181b, miR-181c, miR-181d and miR-127-5p decreased in OA patients. miRNA-127-5p mimics suppressed OPN production as well as the activity of a reporter construct containing the 3'-UTR of human OPN mRNA. In addition, mutation of miR-127-5p binding site in the 3'-UTR of OPN mRNA abolished miR-127-5p-mediated repression of reporter activity. Conversely, treatment with miR-127-5p inhibitor increased reporter activity and OPN production. Interestingly, miR-127-5p inhibited proliferation of chondrocytes through OPN. In conclusion, miRNA-127-5p is an important regulator of OPN in human chondrocytes and may contribute to the development of OA.
Collapse
Affiliation(s)
- Min Tu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410078, China.,Department of Orthopaedics, Second People's Hospital of Jingmen, Jingmen, 448000, China
| | - Yusheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Zhenhan Deng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Shuguang Gao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Wenfeng Xiao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Wei Luo
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Wei Jiang
- Department of Bone and Joint, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzheng, 518020, China
| | - Liangjun Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410078, China.,Department of Joint Surgery, Changsha Central Hospital, Changsha, 410000, China
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410078, China
| |
Collapse
|
28
|
Wu M, Xiao H, Liu G, Chen S, Tan B, Ren W, Bazer FW, Wu G, Yin Y. Glutamine promotes intestinal SIgA secretion through intestinal microbiota and IL-13. Mol Nutr Food Res 2016; 60:1637-48. [PMID: 27005687 DOI: 10.1002/mnfr.201600026] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/02/2016] [Accepted: 02/29/2016] [Indexed: 12/20/2022]
Abstract
SCOPE Glutamine supplementation enhances secretory IgA (SIgA) production in the intestine, but the mechanism is largely unknown. We examined this issue using a mouse model. METHODS AND RESULTS In mouse model, glutamine supplementation increased both SIgA abundance in intestinal luminal contents and IgA(+) plasma cell numbers in the mouse ileum, and decreased bacterial loads in mouse mesenteric lymph nodes. Glutamine supplementation increased mouse ileal expression of cytokines associated with T cell-dependent and T cell-independent pathways of SIgA induction, including IL-5, IL-6, IL-13, transforming growth factor (TGF-β), a proliferation-inducing ligand (APRIL), B cell-activating factor (BAFF), vasoactive intestinal peptide (VIP) receptor, and retinal dehydrogenases. Injecting an IL-13 antibody during glutamine supplementation reduced J-chain expression in the mouse ileum. Glutamine supplementation increased bacterial invasion into the mouse ileal wall, while disrupting the mouse intestinal microbiota abrogated the influence of glutamine supplementation on SIgA secretion. CONCLUSION Glutamine supplementation appears to enhance SIgA secretion in the mouse intestine through the intestinal microbiota and subsequently through T cell-dependent and T cell-independent pathways.
Collapse
Affiliation(s)
- Miaomiao Wu
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China.,Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Hao Xiao
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China
| | - Gang Liu
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China
| | - Shuai Chen
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China
| | - Bie Tan
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China
| | - Wenkai Ren
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX, USA
| | - Yulong Yin
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China
| |
Collapse
|
29
|
Alterations of amino acid metabolism in osteoarthritis: its implications for nutrition and health. Amino Acids 2016; 48:907-914. [DOI: 10.1007/s00726-015-2168-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 12/30/2015] [Indexed: 01/15/2023]
|
30
|
Co-dependence of genotype and dietary protein intake to affect expression on amino acid/peptide transporters in porcine skeletal muscle. Amino Acids 2015; 48:75-90. [PMID: 26255284 DOI: 10.1007/s00726-015-2066-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 07/29/2015] [Indexed: 10/23/2022]
Abstract
A total of 96 barrows (48 pure-bred Bama mini-pigs representing fatty genotype, and 48 Landrace pigs representing lean genotype) were randomly assigned to either a low- or adequate-protein treatment diet. The experimental period commenced at 5 weeks of age and extended to the finishing period. After euthanasia, blood and skeletal muscle samples were collected from pigs at the nursery, growing, and finishing phases. Our results indicate that the concentrations of free AAs in the plasma and muscle decreased as the age of the pigs increased. In addition, a strain × growth phase interaction (P < 0.05) was observed for the free AA pool in the plasma and muscle. The low-protein diet upregulated (P < 0.05) the mRNA levels for T1R1/T1R3 involved in glutamate binding, but downregulated (P < 0.05) the mRNA levels for PAT1, PAT2, and ASCT2, which transport neutral AAs into muscles. Bama mini-pigs had higher (P < 0.05) mRNA levels for LAT1, SNAT2, and EAAC1, but a lower (P < 0.05) mRNA level for PepT1, compared with Landrace pigs. Collectively, our findings indicate that adequate provision of dietary protein plays an important role in regulating profiles of free AA pools and expression of key AA/peptide transporters/transceptors in a genotype- and tissue-specific manner.
Collapse
|
31
|
Duan J, Yin J, Ren W, Liu T, Cui Z, Huang X, Wu L, Kim SW, Liu G, Wu X, Wu G, Li T, Yin Y. Dietary supplementation with L-glutamate and L-aspartate alleviates oxidative stress in weaned piglets challenged with hydrogen peroxide. Amino Acids 2015; 48:53-64. [PMID: 26255283 DOI: 10.1007/s00726-015-2065-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/29/2015] [Indexed: 11/29/2022]
Abstract
This study was to evaluate the protective roles of L-glutamate (Glu) and L-aspartate (Asp) in weaned piglets challenged with H2O2. Forty weaned piglets were assigned randomly into one of five groups (8 piglets/group): (1) control group (NC) in which pigs were fed a corn- and soybean meal-based diet and received intraperitoneal administration of saline; (2) H2O2 group (PC) in which pigs were fed the basal diet and received intraperitoneal administration of 10 % H2O2 (1 ml/kg body weight once on days 8 and repeated on day 11); (3) PC + Glu group (PG) in which pigs were fed the basal diet supplemented with 2.0 % Glu before intraperitoneal administration of 10 % H2O2; (4) PC + Asp group (PA) in which pigs were fed the basal diet supplemented with 1.0 % Asp before intraperitoneal administration of 10 % H2O2; (5) PC + Glu + Asp group (PGA) in which pigs were fed the basal diet supplemented with 2.0 % Glu plus 1.0 % Asp before intraperitoneal administration of 10 % H2O2. Measured parameters included daily feed intake (DFI), average daily gain (ADG), feed conversion rate (FCR), and serum anti-oxidative enzyme activities (catalase, superoxide dismutase, glutathione peroxidase-1), serum malondialdehyde and H2O2 concentrations, serum amino acid (AA) profiles, and intestinal expression of AA transporters. Dietary supplementation with Glu, Asp or their combination attenuated the decreases in DFI, ADG and feed efficiency, the increase in oxidative stress, the alterations of serum AA concentrations, and the changed expression of intestinal AA transporters in H2O2-challenged piglets. Thus, dietary supplementation with Glu or Asp alleviates growth suppression and oxidative stress, while restoring serum the amino acid pool in H2O2-challenged piglets.
Collapse
Affiliation(s)
- Jielin Duan
- 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, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Jie Yin
- 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, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Wenkai Ren
- 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, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Ting Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China
| | - Zhijie Cui
- 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, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Xingguo Huang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China
| | - Li Wu
- 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, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, 27695, USA
| | - Gang Liu
- 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, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Xi Wu
- 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, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Tiejun Li
- 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, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China. .,Changsha Lvye Biotechnology Limited Company Academician Expert Workstation, Changsha, 410128, Hunan, China. .,Guangdong Wangda Group Academician Workstation for Clean Feed Technology Research and Development in Swine, Guangzhou, 510663, Guangdong, China. .,Guangdong Hinapharm Group Academician Workstation for Biological Feed and Feed Additives and Animal Intestinal Health, Guangzhou, 511400, Guangdong, China.
| | - Yulong Yin
- 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, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China. .,School of Biology, Hunan Normal Univesity, Changsha, 410018, Hunan, China.
| |
Collapse
|
32
|
Hou Y, Yin Y, Wu G. Dietary essentiality of "nutritionally non-essential amino acids" for animals and humans. Exp Biol Med (Maywood) 2015; 240:997-1007. [PMID: 26041391 DOI: 10.1177/1535370215587913] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Based on growth or nitrogen balance, amino acids (AA) had traditionally been classified as nutritionally essential (indispensable) or non-essential (dispensable) for animals and humans. Nutritionally essential AA (EAA) are defined as either those AA whose carbon skeletons cannot be synthesized de novo in animal cells or those that normally are insufficiently synthesized de novo by the animal organism relative to its needs for maintenance, growth, development, and health and which must be provided in the diet to meet requirements. In contrast, nutritionally non-essential AA (NEAA) are those AA which can be synthesized de novo in adequate amounts by the animal organism to meet requirements for maintenance, growth, development, and health and, therefore, need not be provided in the diet. Although EAA and NEAA had been described for over a century, there are no compelling data to substantiate the assumption that NEAA are synthesized sufficiently in animals and humans to meet the needs for maximal growth and optimal health. NEAA play important roles in regulating gene expression, cell signaling pathways, digestion and absorption of dietary nutrients, DNA and protein synthesis, proteolysis, metabolism of glucose and lipids, endocrine status, men and women fertility, acid-base balance, antioxidative responses, detoxification of xenobiotics and endogenous metabolites, neurotransmission, and immunity. Emerging evidence indicates dietary essentiality of "nutritionally non-essential amino acids" for animals and humans to achieve their full genetic potential for growth, development, reproduction, lactation, and resistance to metabolic and infectious diseases. This concept represents a new paradigm shift in protein nutrition to guide the feeding of mammals (including livestock), poultry, and fish.
Collapse
Affiliation(s)
- Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yulong Yin
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China
| | - Guoyao Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
33
|
Bujold AR, MacInnes JI. Validation of reference genes for quantitative real-time PCR (qPCR) analysis of Actinobacillus suis. BMC Res Notes 2015; 8:86. [PMID: 25884823 PMCID: PMC4369107 DOI: 10.1186/s13104-015-1045-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 03/04/2015] [Indexed: 12/25/2022] Open
Abstract
Background Quantitative real-time PCR is a valuable tool for evaluating bacterial gene expression. However, in order to make best use of this method, endogenous reference genes for expression data normalisation must first be identified by carefully validating the stability of expression under experimental conditions. Therefore, the objective of this study was to validate eight reference genes of the opportunistic swine pathogen, Actinobacillus suis, grown in aerobic cultures with (Epinephrine) or without (Aerobic) epinephrine in the growth medium and in anoxic static cultures (Anoxic), and sampled during exponential and stationary phases. Results Using the RefFinder tool, expression data were analysed to determine whether comprehensive stability rankings of selected reference genes varied with experimental design. When comparing Aerobic and Epinephrine cultures by growth phase, pyk and rpoB were both among the most stably expressed genes, but when analysing both growth phases together, only pyk remained in the top three rankings. When comparing Aerobic and Anoxic samples, proS ranked among the most stable genes in exponential and stationary phase data sets as well as in combined rankings. When analysing the Aerobic, Epinephrine, and Anoxic samples together, only gyrA ranked consistently among the top three most stably expressed genes during exponential and stationary growth as well as in combined rankings; the rho gene ranked as least stably expressed gene in this data set. Conclusions Reference gene stability should be carefully assessed with the design of the experiment in mind. In this study, even the commonly used reference gene 16S rRNA demonstrated large variability in stability depending on the conditions studied and how the data were analysed. As previously suggested, the best approach may be to use a geometric mean of multiple genes to normalise qPCR results. As researchers continue to validate reference genes for various organisms in multiple growth conditions and sampling time points, it may be possible to make informed predictions as to which genes may be most suitable to validate for a given experimental design, but in the meantime, the reference genes used to normalise qPCR data should be selected with caution.
Collapse
Affiliation(s)
- Adina R Bujold
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Road East, N1G 2W1, Guelph, Ontario, Canada.
| | - Janet I MacInnes
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Road East, N1G 2W1, Guelph, Ontario, Canada.
| |
Collapse
|
34
|
Duan JL, Yin J, Ren WK, Wu MM, Chen S, Cui ZJ, Wu X, Huang RL, Li TJ, Yin YL. Pyrrolidine dithiocarbamate restores gastric damages and suppressive autophagy induced by hydrogen peroxide. Free Radic Res 2015; 49:210-8. [PMID: 25471085 DOI: 10.3109/10715762.2014.993627] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is well known that gastric barrier is very important for protecting host from various insults. Simultaneously, autophagy serving as a prominent cytoprotective and survival pathway under oxidative stress conditions is being increasingly recognized. Thus, this study was conducted for investigating the effect of pyrrolidine dithiocarbamate (PDTC) on gastric barrier function and autophagy under oxidative stress induced by intragastric administration of hydrogen peroxide (H2O2). The gastric tight junction proteins [zonula occludens-1 (ZO1), occludin, and claudin1], autophagic proteins [microtubule-associated protein light chain 3I(LC3I), LC3II, and beclin1], and nuclear factor kappa B (NF-κB) signaling pathway (p65 and IκB kinase α/β) were determined by Western blot. The results showed that H2O2 exposure disturbed gastric barrier function with decreased expression of ZO1, occludin, and claudin1, and reduced gastric autophagy with decreased conversion of LC3I into LC3II in mice. However, treatment with PDTC restored these adverse effects evidenced by increased expression of ZO1 and claudin1 and increased conversion of LC3I into LC3II. Meanwhile, H2O2 exposure decreased normal human gastric epithelial mucosa cell line (GES-1) viability in a concentration-dependent way. However, after being exposed to H2O2, GES-1 exhibited autophagic response which was inconsistent with our in vivo results in mice, while PDTC failed to decrease autophagy in GES-1 induced by H2O2. Simultaneously, the beneficial effect of PDTC on gastric damage and autophagy in mice might be independent of inhibition of NF-κB. In conclusion, PDTC treatment restores gastric damages and reduced autophagy induced by H2O2. Therefore, PDTC may serve as a potential adjuvant therapy for gastric damages.
Collapse
Affiliation(s)
- J L Duan
- 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, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences , Changsha, Hunan , P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kang N, Chen P, Chen Y, Zeng H, He X, Zhu Y. PRMT6 mediates CSE induced inflammation and apoptosis. Int Immunopharmacol 2015; 24:95-101. [PMID: 25481537 DOI: 10.1016/j.intimp.2014.10.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 10/09/2014] [Accepted: 10/28/2014] [Indexed: 01/19/2023]
Abstract
Cigarette smoke extract (CSE) induces apoptosis and inflammation, but the mechanism is unknown. Arginine methyltransferase (PRMT6) catalyzes the asymmetric di-methylation of histone H3 arginine 2 (H3R2me2a) to control global level transcription. We hypothesized that PRMT6 mediates CSE induced apoptosis and inflammation through H3R2me2a. The apoptosis after CSE treatment in human umbilical vein endothelial cells (HUVECs) was fully measured with real-time reverse transcription PCR, western blotting and Annexin-V staining. Meanwhile, the inflammation in HUVECs after CSE exposure was detected with real-time reverse transcription PCR, western blotting and ELISA. CSE treatment promoted apoptosis and inflammation in HUVECs, coinciding with the decreased protein abundance of PRMT6. Meanwhile, HUVECs transfected with PRMT6 expressing plasmid inhibited the CSE-induced apoptosis and inflammation. Also, the inhibition of PRMT6 promoted the apoptosis and inflammation in HUVECs induced by CSE. Notably, H3R2me2a was associated with the modulation of PRMT6 in CSE induced apoptosis and inflammation in HUVECs. In conclusion, PRMT6 mediates CSE induced apoptosis and inflammation through H3R2me2a in HUVECs.
Collapse
Affiliation(s)
- Naixing Kang
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Yan Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China.
| | - Huihui Zeng
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Xue He
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Yingqun Zhu
- Department of Respiratory Medicine, The Third Hospital of Changsha, Changsha, Hunan 410015, China
| |
Collapse
|
36
|
Mouse intestinal innate immune responses altered by enterotoxigenic Escherichia coli (ETEC) infection. Microbes Infect 2014; 16:954-61. [DOI: 10.1016/j.micinf.2014.09.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/04/2014] [Accepted: 09/18/2014] [Indexed: 12/12/2022]
|
37
|
Ren W, Duan J, Yin J, Liu G, Cao Z, Xiong X, Chen S, Li T, Yin Y, Hou Y, Wu G. Dietary L-glutamine supplementation modulates microbial community and activates innate immunity in the mouse intestine. Amino Acids 2014; 46:2403-13. [PMID: 25023447 DOI: 10.1007/s00726-014-1793-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 06/21/2014] [Indexed: 01/08/2023]
Abstract
This study was conducted to determine effects of dietary supplementation with 1 % L-glutamine for 14 days on the abundance of intestinal bacteria and the activation of intestinal innate immunity in mice. The measured variables included (1) the abundance of Bacteroidetes, Firmicutes, Lactobacillus, Streptococcus and Bifidobacterium in the lumen of the small intestine; (2) the expression of toll-like receptors (TLRs), pro-inflammatory cytokines, and antibacterial substances secreted by Paneth cells and goblet cells in the jejunum, ileum and colon; and (3) the activation of TLR4-nuclear factor kappa B (NF-κB), mitogen-activated protein kinases (MAPK), and phosphoinositide-3-kinases (PI3K)/PI3K-protein kinase B (Akt) signaling pathways in the jejunum and ileum. In the jejunum, glutamine supplementation decreased the abundance of Firmicutes, while increased mRNA levels for antibacterial substances in association with the activation of NF-κB and PI3K-Akt pathways. In the ileum, glutamine supplementation induced a shift in the Firmicutes:Bacteroidetes ratio in favor of Bacteroidetes, and enhanced mRNA levels for Tlr4, pro-inflammatory cytokines, and antibacterial substances participating in NF-κB and JNK signaling pathways. These results indicate that the effects of glutamine on the intestine vary with its segments and compartments. Collectively, dietary glutamine supplementation of mice beneficially alters intestinal bacterial community and activates the innate immunity in the small intestine through NF-κB, MAPK and PI3K-Akt signaling pathways.
Collapse
Affiliation(s)
- Wenkai Ren
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, 410125, Hunan, China,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chen S, Liu S, Zhang F, Ren W, Li N, Yin J, Duan J, Peng Y, Liu G, Yin Y, Wu G. Effects of dietary l-glutamine supplementation on specific and general defense responses in mice immunized with inactivated Pasteurella multocida vaccine. Amino Acids 2014; 46:2365-75. [DOI: 10.1007/s00726-014-1789-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 06/13/2014] [Indexed: 10/25/2022]
|
39
|
Wu M, Xiao H, Ren W, Yin J, Tan B, Liu G, Li L, Nyachoti CM, Xiong X, Wu G. Therapeutic effects of glutamic acid in piglets challenged with deoxynivalenol. PLoS One 2014; 9:e100591. [PMID: 24984001 PMCID: PMC4077692 DOI: 10.1371/journal.pone.0100591] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/26/2014] [Indexed: 12/30/2022] Open
Abstract
The mycotoxin deoxynivalenol (DON), one of the most common food contaminants, primarily targets the gastrointestinal tract to affect animal and human health. This study was conducted to examine the protective function of glutamic acid on intestinal injury and oxidative stress caused by DON in piglets. Twenty-eight piglets were assigned randomly into 4 dietary treatments (7 pigs/treatment): 1) uncontaminated control diet (NC), 2) NC+DON at 4 mg/kg (DON), 3) NC+2% glutamic acid (GLU), and 4) NC+2% glutamic acid + DON at 4 mg/kg (DG). At day 15, 30 and 37, blood samples were collected to determine serum concentrations of CAT (catalase), T-AOC (total antioxidant capacity), H2O2 (hydrogen peroxide), NO (nitric oxide), MDA (maleic dialdehyde), DAO (diamine oxidase) and D-lactate. Intestinal morphology, and the activation of Akt/mTOR/4EBP1 signal pathway, as well as the concentrations of H2O2, MDA, and DAO in kidney, liver and small intestine, were analyzed at day 37. Results showed that DON significantly (P<0.05) induced oxidative stress in piglets, while this stress was remarkably reduced with glutamic acid supplementation according to the change of oxidative parameters in blood and tissues. Meanwhile, DON caused obvious intestinal injury from microscopic observations and permeability indicators, which was alleviated by glutamic acid supplementation. Moreover, the inhibition of DON on Akt/mTOR/4EBP1 signal pathway was reduced by glutamic acid supplementation. Collectively, these data suggest that glutamic acid may be a useful nutritional regulator for DON-induced damage manifested as oxidative stress, intestinal injury and signaling inhibition.
Collapse
Affiliation(s)
- Miaomiao Wu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Scienses, Changsha, Hunan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao Xiao
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Scienses, Changsha, Hunan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wenkai Ren
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Scienses, Changsha, Hunan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Yin
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Scienses, Changsha, Hunan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bie Tan
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Scienses, Changsha, Hunan, China
| | - Gang Liu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Scienses, Changsha, Hunan, China
| | - Lili Li
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Scienses, Changsha, Hunan, China
| | | | - Xia Xiong
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Scienses, Changsha, Hunan, China
- * E-mail:
| | - Guoyao Wu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Scienses, Changsha, Hunan, China
- Department of Animal Science, Texas A&M University, College Station, Texas, United State of America
| |
Collapse
|
40
|
Wang B, Wu G, Zhou Z, Dai Z, Sun Y, Ji Y, Li W, Wang W, Liu C, Han F, Wu Z. Glutamine and intestinal barrier function. Amino Acids 2014; 47:2143-54. [PMID: 24965526 DOI: 10.1007/s00726-014-1773-4] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/27/2014] [Indexed: 12/27/2022]
Abstract
The intestinal barrier integrity is essential for the absorption of nutrients and health in humans and animals. Dysfunction of the mucosal barrier is associated with increased gut permeability and development of multiple gastrointestinal diseases. Recent studies highlighted a critical role for glutamine, which had been traditionally considered as a nutritionally non-essential amino acid, in activating the mammalian target of rapamycin cell signaling in enterocytes. In addition, glutamine has been reported to enhance intestinal and whole-body growth, to promote enterocyte proliferation and survival, and to regulate intestinal barrier function in injury, infection, weaning stress, and other catabolic conditions. Mechanistically, these effects were mediated by maintaining the intracellular redox status and regulating expression of genes associated with various signaling pathways. Furthermore, glutamine stimulates growth of the small intestinal mucosa in young animals and also enhances ion transport by the gut in neonates and adults. Growing evidence supports the notion that glutamine is a nutritionally essential amino acid for neonates and a conditionally essential amino acid for adults. Thus, as a functional amino acid with multiple key physiological roles, glutamine holds great promise in protecting the gut from atrophy and injury under various stress conditions in mammals and other animals.
Collapse
Affiliation(s)
- Bin Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China.,Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Zhigang Zhou
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Yuli Sun
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Wei Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Weiwei Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Chuang Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Feng Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China.
| |
Collapse
|
41
|
Wu G. Dietary requirements of synthesizable amino acids by animals: a paradigm shift in protein nutrition. J Anim Sci Biotechnol 2014; 5:34. [PMID: 24999386 PMCID: PMC4082180 DOI: 10.1186/2049-1891-5-34] [Citation(s) in RCA: 190] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 06/08/2014] [Indexed: 12/17/2022] Open
Abstract
Amino acids are building blocks for proteins in all animals. Based on growth or nitrogen balance, amino acids were traditionally classified as nutritionally essential or nonessential for mammals, birds and fish. It was assumed that all the “nutritionally nonessential amino acids (NEAA)” were synthesized sufficiently in the body to meet the needs for maximal growth and optimal health. However, careful analysis of the scientific literature reveals that over the past century there has not been compelling experimental evidence to support this assumption. NEAA (e.g., glutamine, glutamate, proline, glycine and arginine) play important roles in regulating gene expression, cell signaling, antioxidative responses, fertility, neurotransmission, and immunity. Additionally, glutamate, glutamine and aspartate are major metabolic fuels for the small intestine to maintain its digestive function and to protect the integrity of the intestinal mucosa. Thus, diets for animals must contain all NEAA to optimize their survival, growth, development, reproduction, and health. Furthermore, NEAA should be taken into consideration in revising the “ideal protein” concept that is currently used to formulate swine and poultry diets. Adequate provision of all amino acids (including NEAA) in diets enhances the efficiency of animal production. In this regard, amino acids should not be classified as nutritionally essential or nonessential in animal or human nutrition. The new Texas A&M University’s optimal ratios of dietary amino acids for swine and chickens are expected to beneficially reduce dietary protein content and improve the efficiency of their nutrient utilization, growth, and production performance.
Collapse
Affiliation(s)
- Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, Texas 77843, USA
| |
Collapse
|
42
|
Ren W, Chen S, Yin J, Duan J, Li T, Liu G, Feng Z, Tan B, Yin Y, Wu G. Dietary arginine supplementation of mice alters the microbial population and activates intestinal innate immunity. J Nutr 2014; 144:988-95. [PMID: 24670969 DOI: 10.3945/jn.114.192120] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Currently, little is known about the function of arginine in the homeostasis of the intestinal immune system. This study was conducted to test the hypothesis that dietary arginine supplementation may alter intestinal microbiota and innate immunity in mice. Mice were fed a basal diet (containing 0.93% l-arginine; grams per gram) or the basal diet supplemented with 0.5% l-arginine for 14 d. We studied the composition of intestinal microbiota, the activation of innate immunity, and the expression of toll-like receptors (Tlrs), proinflammatory cytokines, and antimicrobials in the jejunum, ileum, or colon of mice. Signal transduction pathway activation in the jejunum and ileum, including TLR4-nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), mitogen-activated protein kinase (MAPK), and phosphoinositide-3 kinase (PI3K)/PI3K-protein kinase B (Akt), was analyzed by Western blotting. Quantitative polymerase chain reaction analysis revealed that arginine supplementation induced (P < 0.05) a shift in the Firmicutes-to-Bacteroidetes ratio to favor Bacteroidetes in the jejunum (0.33 ± 0.04 vs. 1.0 ± 0.22) and ileum (0.20 ± 0.08 vs. 1.0 ± 0.27) compared with the control group. This finding coincided with greater (P < 0.05) activation of the innate immune system, including TLR signaling, as well as expression of proinflammatory cytokines, secretory immunoglobulin A, mucins, and Paneth antimicrobials in the jejunum and ileum. Finally, arginine supplementation reduced (P < 0.05) expression of the proteins for NF-κB, MAPK, and PI3K-Akt signaling pathways but activated (P < 0.05) p38 and c-Jun N-terminal protein kinase in the jejunum and the ileum, respectively. Collectively, dietary arginine supplementation of mice changes the intestinal microbiota, contributing to the activation of intestinal innate immunity through NF-κB, MAPK, and PI3K-phosphorylated Akt signaling pathways.
Collapse
Affiliation(s)
- Wenkai Ren
- 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, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Shuai Chen
- 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, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Jie Yin
- 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, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Jielin Duan
- 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, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Tiejun Li
- 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, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Gang Liu
- 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, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Zemeng Feng
- 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, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Bie Tan
- 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, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Yulong Yin
- 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, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Guoyao Wu
- 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, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and Department of Animal Science, Texas A&M University, College Station, TX
| |
Collapse
|
43
|
Ren W, Yin J, Wu M, Liu G, Yang G, Xion Y, Su D, Wu L, Li T, Chen S, Duan J, Yin Y, Wu G. Serum amino acids profile and the beneficial effects of L-arginine or L-glutamine supplementation in dextran sulfate sodium colitis. PLoS One 2014; 9:e88335. [PMID: 24505477 PMCID: PMC3914992 DOI: 10.1371/journal.pone.0088335] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 01/07/2014] [Indexed: 12/16/2022] Open
Abstract
This study was conducted to investigate serum amino acids profile in dextran sulfate sodium (DSS)-induced colitis, and impacts of graded dose of arginine or glutamine supplementation on the colitis. Using DSS-induced colitis model, which is similar to human ulcerative colitis, we determined serum profile of amino acids at day 3, 7, 10 and 12 (5 days post DSS treatment). Meanwhile, effects of graded dose of arginine (0.4%, 0.8%, and 1.5%) or glutamine (0.5%, 1.0% and 2.0%) supplementation on clinical parameters, serum amino acids, colonic tight junction proteins, colonic anti-oxidative indicators [catalase, total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px)], colonic pro-inflammatory cytokines [interleukin-1 beta (IL-1β), IL-6, IL-17 and tumor necrosis factor alpha (TNF-α)] in DSS-induced colitis were fully analyzed at day 7 and 12. Additionally, the activation of signal transduction pathways, including nuclear factor kappa B (NF-κB), mitogen-activated protein kinases (MAPK), phosphoinositide-3-kinases (PI3K)/PI3K-protein kinase B (Akt), and myosin light chain kinase (MLCK)-myosin light chain (MLC20), were analyzed using immunoblotting. Serum amino acids analysis showed that DSS treatment changed the serum contents of amino acids, such as Trp, Glu, and Gln (P<0.05). Dietary arginine or glutamine supplementation had significant (P<0.05) influence on the clinical and biochemical parameters (T-SOD, IL-17 and TNF-α) in colitis model. These results were associated with colonic NF-κB, PI3K-Akt and MLCK signaling pathways. In conclusion, arginine or glutamine could be a potential therapy for intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Wenkai Ren
- 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 Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Jie Yin
- 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 Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Miaomiao Wu
- 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 Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Gang Liu
- 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 Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Guan Yang
- School of Food Science, Washington State University, Pullman, Washington, United States of America
| | - Yan Xion
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Dingding Su
- Hunan Provincial Key Laboratory for Germplasm Innovation and Utilization of Crop, RuanDa Road# 129, Changsha, Hunan, China
| | - Li Wu
- 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 Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Tiejun Li
- 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 Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Shuai Chen
- 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 Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Jielin Duan
- 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 Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Yulong Yin
- 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 Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Guoyao Wu
- 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 Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
- Department of Animal Science, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|