1
|
Huang Y, Yin C, Wang J, Wang M, Zheng X, Xie M, Wang J. Impact of Glutamine-Enhanced Parenteral Nutrition on Postoperative Outcomes in Colorectal Cancer Patients. Cancer Manag Res 2024; 16:1329-1344. [PMID: 39372706 PMCID: PMC11456277 DOI: 10.2147/cmar.s476648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Purpose This study investigated the effects of parenteral glutamine (Gln) supplement immunonutrition versus conventional nutritional support on postoperative Clavien-Dindo classification complications and recovery, perioperative nutritional status, and immune, inflammation, and safety indicators in patients with colorectal cancer (CRC). Patients and Methods Clinical data were collected for a retrospective cohort study of 178 patients (58 and 120 patients in the observation and control groups, respectively) who underwent radical resection of CRC from January 2019 to December 2021. The incidence of postoperative complications was calculated. Postoperative recovery, nutritional indicators, inflammatory factors indicator, and the safety indicators before operation and at 1, 3, and 7 days after operation were compared. SPSS 29.0 statistical software was used for statistical analysis. Results The incidence of postoperative overall complications in the control group and the observation group was 22.50% (27/120) and 17.24% (10/58), respectively, and there was no significant difference between the two groups (P=0.42). The incidence of postoperative complications of Clavien-Dindo grade ≥III in the control group and the observation group was 14.17% (17/120) and 3.45% (2/58), respectively, and the difference between the two groups was statistically significant (P=0.03). Secondary outcomes (first exhaust, defecation, and liquid diet intake times) were significantly recovered earlier in the observation group than those in the control group (P<0.05), while the postoperative hospital stay was significantly shorter(P=0.04). The perioperative nutritional status did not significantly differ between the groups before and after surgery(P>0.05), although significant differences were observed in several inflammatory and safety indicators(P<0.05). Conclusion Unlike conventional nutritional support, postoperative parenteral Gln supplementation reduced the incidence of postoperative Clavien-Dindo complications grade ≥III in patients with CRC while increasing intestinal and immune functions, decreasing inflammation, and reducing the length of hospital stay.
Collapse
Affiliation(s)
- Yong Huang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Chunmei Yin
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Jue Wang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Maijian Wang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Xingbin Zheng
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Ming Xie
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Jiwei Wang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| |
Collapse
|
2
|
Alanazi AH, Selim MS, Yendamuri MR, Zhang D, Narayanan SP, Somanath PR. The impact of diabetes mellitus on blood-tissue barrier regulation and vascular complications: Is the lung different from other organs? Tissue Barriers 2024:2386183. [PMID: 39072526 DOI: 10.1080/21688370.2024.2386183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024] Open
Abstract
Diabetes Mellitus presents a formidable challenge as one of the most prevalent and complex chronic diseases, exerting significant strain on both patients and the world economy. It is recognized as a common comorbidity among severely ill individuals, often leading to a myriad of micro- and macro-vascular complications. Despite extensive research dissecting the pathophysiology and molecular mechanisms underlying vascular complications of diabetes, relatively little attention has been paid to potential lung-related complications. This review aims to illuminate the impact of diabetes on prevalent respiratory diseases, including chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), idiopathic pulmonary fibrosis (IPF), tuberculosis (TB), pneumonia infections, and asthma, and compare the vascular complications with other vascular beds. Additionally, we explore the primary mechanistic pathways contributing to these complications, such as the expression modulation of blood-tissue-barrier proteins, resulting in increased paracellular and transcellular permeability, and compromised immune responses rendering diabetes patients more susceptible to infections. The activation of inflammatory pathways leading to cellular injury and hastening the onset of these respiratory complications is also discussed.
Collapse
Affiliation(s)
- Abdulaziz H Alanazi
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Department of Clinical Practice, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Mohamed S Selim
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Manyasreeprapti R Yendamuri
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - S Priya Narayanan
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| |
Collapse
|
3
|
Zheng J, Li Y, Kong X, Guo J. Exploring immune-related pathogenesis in lung injury: Providing new insights Into ALI/ARDS. Biomed Pharmacother 2024; 175:116773. [PMID: 38776679 DOI: 10.1016/j.biopha.2024.116773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/08/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) represent a significant global burden of morbidity and mortality, with lung injury being the primary cause of death in affected patients. The pathogenesis of lung injury, however, remains a complex issue. In recent years, the role of the immune system in lung injury has attracted extensive attention worldwide. Despite advancements in our understanding of various lung injury subtypes, significant limitations persist in both prevention and treatment. This review investigates the immunopathogenesis of ALI/ARDS, aiming to elucidate the pathological processes of lung injury mediated by dendritic cells (DCs), natural killer (NK) cells, phagocytes, and neutrophils. Furthermore, the article expounds on the critical contributions of gut microbiota, inflammatory pathways, and cytokine storms in the development of ALI/ARDS.
Collapse
Affiliation(s)
- Jiajing Zheng
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Li
- Pharmacy Department of the First Affiliated Hospital, Henan University of Science and Technology, Luoyang 471000, China
| | - Xianbin Kong
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Jinhe Guo
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
4
|
Ji X, Ji HL. Metabolic signatures of acute respiratory distress syndrome: COVID versus non-COVID. Am J Physiol Lung Cell Mol Physiol 2024; 326:L596-L603. [PMID: 38469648 PMCID: PMC11380973 DOI: 10.1152/ajplung.00266.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 03/13/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a fatal pulmonary disorder characterized by severe hypoxia and inflammation. ARDS is commonly triggered by systemic and pulmonary infections, with bacteria and viruses. Notable pathogens include Pseudomonas aeruginosa, Streptococcus aureus, Enterobacter species, coronaviruses, influenza viruses, and herpesviruses. COVID-19 ARDS represents the latest etiological phenotype of the disease. The pathogenesis of ARDS caused by bacteria and viruses exhibits variations in host immune responses and lung mesenchymal injury. We postulate that the systemic and pulmonary metabolomics profiles of ARDS induced by COVID-19 pathogens may exhibit distinctions compared with those induced by other infectious agents. This review aims to compare metabolic signatures in blood and lung specimens specifically within the context of ARDS. Both prevalent and phenotype-specific metabolomic signatures, including but not limited to glycolysis, ketone body production, lipid oxidation, and dysregulation of the kynurenine pathways, were thoroughly examined in this review. The distinctions in metabolic signatures between COVID-19 and non-COVID ARDS have the potential to reveal new biomarkers, elucidate pathogenic mechanisms, identify druggable targets, and facilitate differential diagnosis in the future.
Collapse
Affiliation(s)
- Xiangming Ji
- Department of Nutrition, Georgia State University, Atlanta, Georgia, United States
| | - Hong-Long Ji
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, Illinois, United States
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, Illinois, United States
| |
Collapse
|
5
|
Ex Vivo Evaluation of Glutamine Treatment in Sepsis and Trauma in a Human Peripheral Blood Mononuclear Cells Model. Nutrients 2023; 15:nu15010252. [PMID: 36615909 PMCID: PMC9824313 DOI: 10.3390/nu15010252] [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: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023] Open
Abstract
We aimed to assess the lipopolysaccharide (LPS), or heat shock (HS) induction, and glutamine-modulating effects on heat shock protein-90α (HSP90α) and cytokines in an ex vivo model using peripheral blood mononuclear cells (PBMCs). The PBMCs of patients with septic shock, trauma-related systemic inflammatory response syndrome (SIRS), and healthy subjects were incubated with 1 μg/mL LPS at 43 °C (HS). Glutamine 10 mM was added 1 hour before or after induction or not at all. We measured mRNA HSP90α, monocyte (m) and lymphocyte (l) HSP90α proteins, interleukin (IL)-1b, -6, -8, -10, tumor necrosis factor-α (TNF-α), and monocyte chemoattractant protein-1 (MCP-1) supernatant levels. Heat shock increased the HSP90α mRNA and mHSP90α in all groups (10-fold in sepsis, p < 0.001 and p = 0.047, respectively). LPS induced the mHSP90α and lHSP90α in healthy (p < 0.001) and mHSP90α in SIRS (p = 0.004) but not in sepsis. LPS induced the cytokines at 24 and 48 h in all groups, especially in trauma (p < 0.001); HS only induced the IL-8 in healthy (p = 0.003) and septic subjects (p = 0.05). Glutamine at 10 mM before or after stimulation did not alter any induction effect of LPS or HS on HSP90α mRNA and mHSP90α protein in sepsis. In SIRS, glutamine before LPS decreased the mHSP90α but increased it when given after HS (p = 0.018). Before or after LPS (p = 0.049) and before HS (p = 0.018), glutamine decreased the lHSP90α expression in sepsis but increased it in SIRS when given after HS (p = 0.003). Regarding cytokines, glutamine enhanced the LPS-induced MCP-1 at 48 h in healthy (p = 0.011), SIRS (p < 0.001), and sepsis (p = 0.006). In conclusion, glutamine at 10 mM, before or after LPS and HS, modulates mHSP90α and lHSP90α in sepsis and SIRS differently and unpredictably. Although it does not alter the stimulation effect on interleukins, glutamine enhances the LPS induction effect on supernatant MCP-1 in all groups. Future research should seek to elucidate better the impact of glutamine and temperature modulation on HSP90α and MCP-1 pathways in sepsis and trauma.
Collapse
|
6
|
Yang M, Zhang X, Zhao S, Shao R, Fan K, Hu K, Zhang L, Yang Y. Protective effects of glutamine on lipopolysaccharide/D-galactosamine-induced fulminant hepatitis in mice. Exp Biol Med (Maywood) 2023; 248:70-78. [PMID: 36259626 PMCID: PMC9989145 DOI: 10.1177/15353702221126562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Fulminant hepatitis remains a critical health problem owing to its high mortality rate and the lack of effective therapies. An increasing number of studies have shown that glutamine supplementation provides protective benefits in inflammation-related disorders, but the pharmacological significance of glutamine in lipopolysaccharide (LPS)/D-galactosamine (D-Gal)-induced fulminant hepatitis remains unclear. In the present study, the potential effects of glutamine on LPS/D-Gal-induced fulminant hepatitis were investigated. Pretreatment with glutamine decreased plasma activities of alanine and aspartate aminotransferases, and ameliorated hepatic morphological abnormalities in LPS/D-Gal-exposed mice. Glutamine pretreatment also inhibited LPS/D-Gal-induced tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6) production. In addition, glutamine pretreatment decreased the level of cleaved cysteinyl aspartate-specific proteinase 3 (caspase-3), suppressed the activities of caspase-3, caspase-8, and caspase-9, and reduced the number of cells positive for TdT-mediated dUTP nick-end labeling in LPS/D-Gal-challenged mice. Interestingly, post-treatment with glutamine also provided protective benefits against LPS/D-Gal-induced acute liver injury, although these effects were less robust than those of glutamine pre-treatment. Thus, glutamine may have potential value as a pharmacological intervention in fulminant hepatitis.
Collapse
Affiliation(s)
- Mengxin Yang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Xinyue Zhang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Shuang Zhao
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Ruyue Shao
- Clinical Medical School, Chongqing Medical and Pharmaceutical College, Chongqing 400016, China
| | - Kerui Fan
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Kai Hu
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Li Zhang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yongqiang Yang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
7
|
Recomendaciones para la terapia nutricional de pacientes con COVID-19. ACTA COLOMBIANA DE CUIDADO INTENSIVO 2021. [PMCID: PMC7843081 DOI: 10.1016/j.acci.2021.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
8
|
Glutamine Administration Attenuates Kidney Inflammation in Obese Mice Complicated with Polymicrobial Sepsis. Mediators Inflamm 2021; 2021:5597118. [PMID: 33859538 PMCID: PMC8024070 DOI: 10.1155/2021/5597118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/04/2021] [Accepted: 03/12/2021] [Indexed: 11/17/2022] Open
Abstract
Obesity is a well-known public health issue around the world. Sepsis is a lethal clinical syndrome that causes multiorgan failure. Obesity may aggravate inflammation in septic patients. Glutamine (GLN) is a nutrient with immune regulatory and anti-inflammatory properties. Since sepsis is a common contributing factor for acute kidney injury (AKI), this study investigated the effects of GLN administration on sepsis-induced inflammation and AKI in obese mice. A high-fat diet which consists of 60% of calories from fat was provided for 10 weeks to induce obesity in the mice. Then, the obese mice were subdivided into sepsis with saline (SS) or GLN (SG) groups. Cecal ligation and puncture (CLP) was performed to produce sepsis. The SS group was intravenously injected with saline while the SG group was administered GLN one or two doses after CLP. Obese mice with sepsis were sacrificed at 12, 24, or 48 h post-CLP. Results revealed that sepsis resulted in upregulated high-mobility group box protein-1 pathway-associated gene expression in obese mice. Also, expressions of macrophage/neutrophil infiltration markers and inflammatory cytokines in kidneys were elevated. Obese mice treated with GLN after sepsis reversed the depletion of plasma GLN, reduced production of lipid peroxides, and downregulated macrophage/neutrophil infiltration and the inflammatory-associated pathway whereas tight junction gene expression increased in the kidneys. These findings suggest that intravenously administered GLN to obese mice after sepsis alleviated inflammation and attenuated AKI. This model may have clinical application to obese patients with a risk for infection in abdominal surgery.
Collapse
|
9
|
Cheng X, Aabdin ZU, Wang Y, Ma N, Dai H, Shi X, Shen X. Glutamine pretreatment protects bovine mammary epithelial cells from inflammation and oxidative stress induced by γ-d-glutamyl-meso-diaminopimelic acid (iE-DAP). J Dairy Sci 2020; 104:2123-2139. [PMID: 33358155 DOI: 10.3168/jds.2020-18402] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022]
Abstract
Glutamine (GLN) has many types of biological activity in rats, including anti-inflammatory, antioxidative stress, and anti-apoptosis effects. However, little is known about the effects of GLN on bovine mammary epithelial cells (BMEC). γ-d-Glutamyl-meso-diaminopimelic acid (iE-DAP) is a cell wall peptidoglycan component of gram-negative bacteria that can be recognized by the intracellular receptor nucleotide-binding oligomerization domain-containing protein 1 (NOD1) and can cause bovine mastitis. The goal of the present study was to investigate whether GLN protects BMEC from iE-DAP-induced inflammation, oxidative stress, and apoptosis. We cultured BMEC in a GLN-free medium for 24 h and then separated them into 4 groups: cells treated with 1× PBS for 26 or 32 h (control); cells stimulated by 10 μg/mL iE-DAP for 2 or 8 h (2- or 8-h iE-DAP); cells pretreated with 8 or 4 mM GLN for 24 h followed by 2 or 8 h of 1× PBS treatment (8 or 4 mM GLN); and cells pretreated with 8 or 4 mM GLN for 24 h followed by 2 or 8 h of iE-DAP treatment (DG). In the 2-h iE-DAP group, when levels of inflammation peaked, iE-DAP treatment increased both the mRNA and protein expression of NOD1, inhibitor of nuclear factor-κB (NFKBIA, IκB), and nuclear factor-κB subunit p65 (RELA, NF-κB p65), as well as the mRNA expression of IL6 and IL8 and levels of IL-6 and tumor necrosis factor-α in cell culture supernatants. In contrast, 8 mM GLN pretreatment inhibited the mRNA and protein expression of inflammatory-related factors by suppressing the NOD1/NF-κB pathway. In the 8-h iE-DAP group, iE-DAP treatment decreased the mRNA and protein expression of extracellular regulated kinase (Erk, ERK) and nuclear factor erythroid 2-associated factor2 (NFE2L2, Nrf2), as well as the mRNA expression of superoxide dismutase 1 (SOD1), catalase (CAT), coenzyme II oxidoreductase 1 (NQO1), and heme oxygenase 1 (HMOX1, HO1). In addition, iE-DAP treatment increased the expression of malondialdehyde in BMEC when oxidative stress levels peaked. Interestingly, 4 mM GLN pretreatment induced the mRNA and protein expression of antioxidative stress-related factors and inhibited the expression of reactive oxygen species in BMEC by promoting the ERK/Nrf2 pathway. Moreover, GLN reduced apoptosis caused by inflammation and oxidative stress in BMEC. This is the first report showing that GLN protects against iE-DAP-induced inflammation and oxidative stress via the NOD1/NF-κB and ERK/Nrf2 pathways in BMEC.
Collapse
Affiliation(s)
- Xiaoye Cheng
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zain-Ui Aabdin
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yan Wang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Nana Ma
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Hongyu Dai
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoli Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangzhen Shen
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
10
|
Effects of the Glutamine Administration on T Helper Cell Regulation and Inflammatory Response in Obese Mice Complicated with Polymicrobial Sepsis. Mediators Inflamm 2020; 2020:8869017. [PMID: 33223959 PMCID: PMC7671796 DOI: 10.1155/2020/8869017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
This study investigated the impacts of GLN on inflammation and T cell dysregulation in obese mice complicated with sepsis. Mice were divided into normal control (NC) and high-fat diet groups. The high-fat diet provided 60% of energy from fat and was administered for 10 weeks to induce obesity. Mice fed with a high-fat diet were then assigned to sham (SH) and sepsis with saline (SS) or GLN (SG) groups. The SH group was subjected to laparotomy, while the sepsis group underwent cecal ligation and puncture (CLP). The SS group was intravenously injected with saline. The SG group was intravenously administered GLN after CLP. Mice were sacrificed at 12, 24, or 48 h post-CLP, respectively. Results demonstrated that in the presence of obesity, sepsis drove CD4+ T cells toward the helper T (Th)2 and Th17 lineages. Also, expressions of inflammatory cytokines and macrophage infiltration markers in adipose tissues and lungs were elevated. Treatment of obese mice with GLN after sepsis reversed Th polarization and downregulated macrophage infiltration and inflammatory cytokine, whereas the tight junction-associated protein expression increased in the lungs. These findings suggest that the intravenous administration of GLN to obese mice after sepsis modulated a more balanced Th cell lineage, alleviated inflammation, and attenuated lung injury.
Collapse
|
11
|
Sahebnasagh A, Saghafi F, Safdari M, Khataminia M, Sadremomtaz A, Talaei Z, Rezai Ghaleno H, Bagheri M, Habtemariam S, Avan R. Neutrophil elastase inhibitor (sivelestat) may be a promising therapeutic option for management of acute lung injury/acute respiratory distress syndrome or disseminated intravascular coagulation in COVID-19. J Clin Pharm Ther 2020; 45:1515-1519. [PMID: 32860252 DOI: 10.1111/jcpt.13251] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/30/2020] [Accepted: 07/30/2020] [Indexed: 12/15/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE This article summarizes the effects of sivelestat on acute lung injury/acute respiratory distress syndrome (ALI/ARDS) or ARDS with coagulopathy, both of which are frequently seen in patients with COVID-19. COMMENT COVID-19 patients are more susceptible to thromboembolic events, including disseminated intravascular coagulation (DIC). Various studies have emphasized the role of neutrophil elastase (NE) in the development of DIC in patients with ARDS and sepsis. It has been shown that NE inhibition by sivelestat mitigates ALI through amelioration of injuries in alveolar epithelium and vascular endothelium, as well as reversing the neutrophil-mediated increased vascular permeability. WHAT IS NEW AND CONCLUSIONS Sivelestat, a selective NE inhibitor, has not been evaluated for its possible therapeutic effects against SARS-CoV-2 infection. Based on its promising beneficial effects in underlying complications of COVID-19, sivelestat could be considered as a promising modality for better management of COVID-19-induced ALI/ARDS or coagulopathy.
Collapse
Affiliation(s)
- Adeleh Sahebnasagh
- Department of Internal Medicine, Clinical Research Center, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Fatemeh Saghafi
- Department of Clinical Pharmacy, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammadreza Safdari
- Department of Orthopedic Surgery, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Masoud Khataminia
- Student Research Committee, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Afsaneh Sadremomtaz
- XB20 Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Zeinab Talaei
- Department of Chemistry, Faculty of Science, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Hassan Rezai Ghaleno
- Department of Surgery, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mahdi Bagheri
- Baqiyatallah Hospital, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories and Herbal Analysis Services, School of Science, University of Greenwich, Kent, UK
| | - Razieh Avan
- Department of Clinical Pharmacy, Medical Toxicology and Drug Abuse Research Center (MTDRC), Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
12
|
Dzobo K, Chiririwa H, Dandara C, Dzobo W. Coronavirus Disease-2019 Treatment Strategies Targeting Interleukin-6 Signaling and Herbal Medicine. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 25:13-22. [PMID: 32857671 DOI: 10.1089/omi.2020.0122] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Coronavirus disease-2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is evolving across the world and new treatments are urgently needed as with vaccines to prevent the illness and stem the contagion. The virus affects not only the lungs but also other tissues, thus lending support to the idea that COVID-19 is a systemic disease. The current vaccine and treatment development strategies ought to consider such systems medicine perspectives rather than a narrower focus on the lung infection only. COVID-19 is associated with elevated levels of the inflammatory cytokines such as interleukin-6 (IL-6), IL-10, and interferon-gamma (IFN-γ). Elevated levels of cytokines and the cytokine storm have been linked to fatal disease. This suggests new therapeutic strategies through blocking the cytokine storm. IL-6 is one of the major cytokines associated with the cytokine storm. IL-6 is also known to display pleiotropic/diverse pathophysiological effects. We suggest the blockage of IL-6 signaling and its downstream mediators such as Janus kinases (JAKs), and signal transducer and activators of transcription (STATs) offer potential hope for the treatment of severe cases of COVID-19. Thus, repurposing of already approved IL-6-JAK-STAT signaling inhibitors as well as other anti-inflammatory drugs, including dexamethasone, is under development for severe COVID-19 cases. We conclude this expert review by highlighting the potential role of precision herbal medicines, for example, the Cannabis sativa, provided that omics technologies can be utilized to build a robust scientific evidence base on their clinical safety and efficacy. Precision herbal medicine buttressed by omics systems science would also help identify new molecular targets for drug discovery against COVID-19.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Harry Chiririwa
- Department of Chemical Engineering, Vaal University of Technology, Vanderbijlpark, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Witness Dzobo
- Immunology Department, Pathology, University Hospital Southampton, Southampton, United Kingdom.,Faculty of Science, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
13
|
Heck T, Ludwig M, Frizzo M, Rasia-Filho A, Homem de Bittencourt PI. Suppressed anti-inflammatory heat shock response in high-risk COVID-19 patients: lessons from basic research (inclusive bats), light on conceivable therapies. Clin Sci (Lond) 2020; 134:1991-2017. [PMID: 32749472 PMCID: PMC7403894 DOI: 10.1042/cs20200596] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/05/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
The major risk factors to fatal outcome in COVID-19 patients, i.e., elderliness and pre-existing metabolic and cardiovascular diseases (CVD), share in common the characteristic of being chronic degenerative diseases of inflammatory nature associated with defective heat shock response (HSR). The molecular components of the HSR, the principal metabolic pathway leading to the physiological resolution of inflammation, is an anti-inflammatory biochemical pathway that involves molecular chaperones of the heat shock protein (HSP) family during homeostasis-threatening stressful situations (e.g., thermal, oxidative and metabolic stresses). The entry of SARS coronaviruses in target cells, on the other hand, aggravates the already-jeopardized HSR of this specific group of patients. In addition, cellular counterattack against virus involves interferon (IFN)-mediated inflammatory responses. Therefore, individuals with impaired HSR cannot resolve virus-induced inflammatory burst physiologically, being susceptible to exacerbated forms of inflammation, which leads to a fatal "cytokine storm". Interestingly, some species of bats that are natural reservoirs of zoonotic viruses, including SARS-CoV-2, possess an IFN-based antiviral inflammatory response perpetually activated but do not show any sign of disease or cytokine storm. This is possible because bats present a constitutive HSR that is by far (hundreds of times) more intense and rapid than that of human, being associated with a high core temperature. Similarly in humans, fever is a physiological inducer of HSR while antipyretics, which block the initial phase of inflammation, impair the resolution phase of inflammation through the HSR. These findings offer a rationale for the reevaluation of patient care and fever reduction in SARS, including COVID-19.
Collapse
Affiliation(s)
- Thiago Gomes Heck
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS), Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
| | - Mirna Stela Ludwig
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS), Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
| | - Matias Nunes Frizzo
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS), Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
| | - Alberto Antonio Rasia-Filho
- Federal University of Health Sciences of Porto Alegre (UFCSPA), Graduate Program in Biosciences, Porto Alegre, RS, 90050-170 Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology, Department of Physiology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, 90050-170 Brazil
| |
Collapse
|
14
|
Antecedent Dietary Glutamine Supplementation Benefits Modulation of Liver Pyroptosis in Mice with Polymicrobial Sepsis. Nutrients 2020; 12:nu12041086. [PMID: 32295272 PMCID: PMC7230693 DOI: 10.3390/nu12041086] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022] Open
Abstract
The liver is the main organ responsible for bacterial and endotoxin clearance. Pyroptosis is a form of proinflammatory programmed cell death activated by caspase-1/11 and gasdermin D (GadD). Pyroptosis protects the host against bacterial infection; however, overactive pyroptosis can lead to organ injury. Glutamine (GLN) is a specific amino acid with anti-inflammatory and immunomodulatory properties. This study investigated the effects of GLN pretreatment on liver pyroptosis in a mouse model of polymicrobial sepsis. Mice were assigned to sham, sepsis control (Sepsis-C), and sepsis GLN (Sepsis-G) groups. The sham and Sepsis-C groups were fed the AIN-93G diet. The Sepsis-G group was provided with identical diet components except that part of the casein was replaced by GLN. After feeding the respective diets for 2 weeks, a cecal ligation and puncture (CLP) procedure was performed in the sepsis groups. An antibiotic was administered after CLP. Mice were sacrificed at either 24 or 72 h after CLP. The results showed that sepsis resulted in upregulated liver caspase-1/11 expression. Compared to the Sepsis-C group, the Sepsis-G group had higher liver caspase-11 and NLRP3 gene expressions at 24 h and lower active caspase-1/11 and cleaved GadD protein levels at 72 h after sepsis. Additionally, liver inflammatory cytokine gene expressions had decreased by 72 h post-CLP. The findings suggest that prophylactic administration of GLN initially upregulated liver pyroptosis to eradicate pathogens, yet the process of pyroptosis was suppressed in the late phase of sepsis. This may have beneficially attenuated liver inflammation and injury in an antibiotic-treated septic condition.
Collapse
|
15
|
Abstract
There is abundant evidence that infectious sepsis both in humans and mice with polymicrobial sepsis results in robust activation of complement. Major complement activation products involved in sepsis include C5a anaphylatoxin and its receptors (C5aR1 and C5aR2) and, perhaps, the terminal complement activation product, C5b-9. These products (and others) also cause dysfunction of the innate immune system, with exaggerated early proinflammatory responses, followed by decline of the innate immune system, leading to immunosuppression and multiorgan dysfunction. Generation of C5a during sepsis also leads to activation of neutrophils and macrophages and ultimate appearance of extracellular histones, which have powerful proinflammatory and prothrombotic activities. The distal complement activation product, C5b-9, triggers intracellular Ca fluxes in epithelial and endothelial cells. Histones activate the NLRP3 inflammasome, products of which can damage cells. C5a also activates MAPKs and Akt signaling pathways in cardiomyocytes, causing buildup of [Ca]i, defective action potentials and substantial cell dysfunction, resulting in cardiac and other organ dysfunction. Cardiac dysfunction can be quantitated by ECHO-Doppler parameters. In vivo interventions that block these complement-dependent products responsible for organ dysfunction in sepsis reduce the intensity of sepsis. The obvious targets in sepsis are C5a and its receptors, histones, and perhaps the MAPK pathways. Blockade of C5 has been considered in sepsis, but the FDA-approved antibody (eculizumab) is known to compromise defenses against neisseria and pneumonococcal bacteria, and requires immunization before the mAb to C5 can be used clinically. Small molecular blocking agents for C5aRs are currently in development and may be therapeutically effective for treatment of sepsis.
Collapse
|
16
|
de Fatima Silva F, de Morais H, Ortiz Silva M, da Silva FG, Vianna Croffi R, Serrano-Nascimento C, Rodrigues Graciano MF, Rafael Carpinelli A, Barbosa Bazotte R, de Souza HM. Akt activation by insulin treatment attenuates cachexia in Walker-256 tumor-bearing rats. J Cell Biochem 2020; 121:4558-4568. [PMID: 32056265 DOI: 10.1002/jcb.29682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/16/2020] [Indexed: 11/11/2022]
Abstract
Cancer-bearing often exhibits hypoinsulinemia, insulin (INS) resistance and glutamine depletion associated with cachexia. However, INS and glutamine effects on cachexia metabolic abnormalities, particularly on tumor-affected proteins related to INS resistance, are poorly known. The main purpose of this study was to investigate the effects of INS and glutamine dipeptide (GDP) treatments on phospho-protein kinase B (p-Akt), and phospho-hormone sensitive lipase (p-HSL) in Walker-256 tumor-bearing rats. INS (NPH, 40 UI/kg, subcutaneous), GDP (1.5 g/kg, oral), INS+GDP or vehicle (control rats) were administered for 13 days, once a day, starting at the day of inoculation of tumor cells. The experiments were performed 4 hours after the last treatment to evaluate acute effects of INS and GDP, besides the chronic effects. INS and/or INS+GDP treatments, which markedly increased the insulinemia, increased the p-Akt: total Akt ratio and prevented the increased p-HSLSer552 : total HSL ratio in the retroperitoneal fat of tumor-bearing rats, without changing the INS resistance and increased expression of factor tumor necrosis-α (TNF-α) in this tissue. INS and INS+GDP also increased the p-Akt: total Akt ratio, whereas GDP and INS+GDP increased the GLUT4 glucose transporter gene expression, in the gastrocnemius muscle of the tumor-bearing rats. Accordingly, treatments with INS and INS+GDP markedly reduced glycemia, increased retroperitoneal fat and attenuated the body mass loss of tumor-bearing rats. In conclusion, hyperinsulinemia induced by high-dose INS treatments increased Akt phosphorylation and prevented increased p-HSLSer552 : total HSL ratio, overlapping INS resistance. These effects are consistent with increased fat mass gain and weight loss (cachexia) attenuation of tumor-bearing rats, evidencing that Akt activation is a potential strategy to prevent loss of fat mass in cancer cachexia.
Collapse
Affiliation(s)
| | - Hely de Morais
- Department of Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
| | - Milene Ortiz Silva
- Department of Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
| | | | - Rafael Vianna Croffi
- Department of Physiology and Biophysics, University of São Paulo, São Paulo, Parana, Brazil
| | | | | | | | - Roberto Barbosa Bazotte
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringa, Parana, Brazil
| | - Helenir Medri de Souza
- Department of Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
| |
Collapse
|
17
|
Gameiro PA, Struhl K. Nutrient Deprivation Elicits a Transcriptional and Translational Inflammatory Response Coupled to Decreased Protein Synthesis. Cell Rep 2020; 24:1415-1424. [PMID: 30089253 PMCID: PMC6419098 DOI: 10.1016/j.celrep.2018.07.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 05/22/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022] Open
Abstract
Nutrient deprivation inhibits mRNA translation through mTOR and eIF2α signaling, but it is unclear how the translational program is controlled to reflect the degree of a metabolic stress. In a model of breast cellular transformation, various forms of nutrient deprivation differentially affect the rate of protein synthesis and its recovery over time. Genome-wide translational profiling of glutamine-deprived cells reveals a rapid upregulation of mRNAs containing uORFs and downregulation of ribosomal protein mRNAs, which are followed by selective translation of cytokine and inflammatory mRNAs. Transcription and translation of inflammatory and cytokine genes are stimulated in response to diverse metabolic stresses and depend on eIF2α phosphorylation, with the extent of stimulation correlating with the decrease in global protein synthesis. In accord with the inflammatory stimulus, glutamine deprivation stimulates the migration of transformed cells. Thus, pro-inflammatory gene expression is coupled to metabolic stress, and this can affect cancer cell behavior upon nutrient limitation. Deprivation of some nutrients may impose more constraints on mRNA translation than others. Gameiro and Struhl describe a relationship between translational repression and pro-inflammatory gene expression in response to various metabolic stresses. The pro-inflammatory transcriptional and translational response is not triggered by mTOR inhibition, per se, and requires eIF2α phosphorylation.
Collapse
Affiliation(s)
- Paulo A Gameiro
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Kevin Struhl
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
18
|
Petry ÉR, Dresch DDF, Carvalho C, Medeiros PC, Rosa TG, de Oliveira CM, Martins LAM, Schemitt E, Bona S, Guma FCR, Marroni NP, Wannmacher CMD. Oral glutamine supplementation attenuates inflammation and oxidative stress-mediated skeletal muscle protein content degradation in immobilized rats: Role of 70 kDa heat shock protein. Free Radic Biol Med 2019; 145:87-102. [PMID: 31505269 DOI: 10.1016/j.freeradbiomed.2019.08.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/19/2019] [Accepted: 08/30/2019] [Indexed: 12/19/2022]
Abstract
Skeletal muscle disuse results in myofibrillar atrophy and protein degradation, via inflammatory and oxidative stress-mediated NF-kB signaling pathway activation. Nutritional interventions, such as l-glutamine (GLN) supplementation have shown antioxidant properties and cytoprotective effects through the modulation on the 70-kDa heat shock protein (HSP70) expression. However, these GLN-mediated effects on cell signaling pathways and biochemical mechanisms that control the myofibrillar protein content degradation in muscle disuse situations are poorly known yet. This study investigated the effects of oral GLN plus l-alanine (ALA; GLN + ALA-solution) supplementation, either in their free or dipeptide (L-alanyl-l-glutamine-DIP) form, on GLN-glutathione (GSH) axis and cytoprotection mediated by HSP70 protein expression in the slow-twitch soleus and fast-twitch gastrocnemius skeletal muscle of rats submitted to 14-days of hindlimb immobilization-induced disuse muscle atrophy. Forty-eight Wistar rats were distributed into 6 groups: hindlimb immobilized (IMOB group) and hindlimb immobilized orally supplemented with either GLN (1 g kg-1) plus ALA (0.61 g kg-1) (GLN + ALA-IMOB group) or 1.49 g kg-1 of DIP (DIP-IMOB group) and; no-immobilized (CTRL) and no-immobilized supplemented GLN + ALA and DIP baselines groups. All animals, including CTRL and IMOB rats (water), were supplemented via intragastric gavage for 14 days, concomitantly to immobilization period. Plasma and muscle GLN levels, lipid (thiobarbituric acid reactive substances-TBARS) and protein (carbonyl) peroxidation, erythrocyte concentration of reduced GSH and GSH disulfide (GSSG), plasma and muscle pro-inflammatory TNF-α levels, muscle IKKα/β-NF-kB signaling pathway and, the myofibrillar protein content (MPC) were measured. The MPC was significantly lower in IMOB rats, compared to CTRL, GLN + ALA, and DIP animals (p < 0.05). This finding was associated with reduced plasma and muscle GLN concentration, equally in IMOB animals. Conversely, both GLN + ALA and DIP supplementation restored plasma and muscle GLN levels, which equilibrated GSH and intracellular redox status (GSSG/GSH ratio) in erythrocytes and skeletal muscle even as, increased muscle HSP70 protein expression; attenuating oxidative stress and TNF-α-mediated NF-kB pathway activation, fact that reverberated on reduction of MPC degradation in GLN + ALA-IMOB and DIP-IMOB animals (p < 0.05). In conclusion, the findings shown herein support the oral GLN + ALA and DIP supplementations as a therapeutic and effective nutritional alternative to attenuate the deleterious effects of the skeletal muscle protein degradation induced by muscle disuse.
Collapse
Affiliation(s)
- Éder Ricardo Petry
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Department of Biochemistry, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil.
| | | | - Clarice Carvalho
- Department of Biochemistry, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Tatiana Gomes Rosa
- Famaqui - Mario Quintana Faculty, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cleverson Morais de Oliveira
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Department of Biochemistry, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Leo Anderson Meira Martins
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Department of Biochemistry, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Elizangêla Schemitt
- Post-Graduate Program in Medicine: Medical Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil; Laboratory of Experimental Hepatology and Gastroenterology, Experimental Research Center, Clinical Hospital of Porto Alegre (HCPA), UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Silvia Bona
- Post-Graduate Program in Medicine: Medical Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil; Laboratory of Experimental Hepatology and Gastroenterology, Experimental Research Center, Clinical Hospital of Porto Alegre (HCPA), UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fátima Costa Rodrigues Guma
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Department of Biochemistry, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Norma Possas Marroni
- Post-Graduate Program in Medicine: Medical Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil; Laboratory of Experimental Hepatology and Gastroenterology, Experimental Research Center, Clinical Hospital of Porto Alegre (HCPA), UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Clóvis Milton Duval Wannmacher
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Department of Biochemistry, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
19
|
Vigeland CL, Beggs HS, Collins SL, Chan‐Li Y, Powell JD, Doerschuk CM, Horton MR. Inhibition of glutamine metabolism accelerates resolution of acute lung injury. Physiol Rep 2019; 7:e14019. [PMID: 30821123 PMCID: PMC6395309 DOI: 10.14814/phy2.14019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 02/09/2019] [Indexed: 12/18/2022] Open
Abstract
Despite recent advances, acute respiratory distress syndrome (ARDS) remains a severe and often fatal disease for which there is no therapy able to reduce the underlying excessive lung inflammation or enhance resolution of injury. Metabolic programming plays a critical role in regulating inflammatory responses. Due to their high metabolic needs, neutrophils, macrophages, and lymphocytes rely upon glutamine metabolism to support activation and function. Additionally, during times of physiologic stress, nearly all cells, including fibroblasts and epithelial cells, require glutamine metabolism. We hypothesized that inhibiting glutamine metabolism reduces lung inflammation and promotes resolution of acute lung injury. Lung injury was induced by instilling lipopolysaccharide (LPS) intratracheally. To inhibit glutamine metabolism, we administered a glutamine analogue, 6-diazo-5-oxo-L-norleucine (DON) that binds to glutamine-utilizing enzymes and transporters, after injury was well established. Treatment with DON led to less lung injury, fewer lung neutrophils, lung inflammatory and interstitial macrophages, and lower levels of proinflammatory cytokines and chemokines at 5 and/or 7 days after injury. Additionally, DON led to earlier expression of the growth factor amphiregulin and more rapid recovery of LPS-induced weight loss. Thus, DON reduced lung inflammation and promoted resolution of injury. These data contribute to our understanding of how glutamine metabolism regulates lung inflammation and repair, and identifies a novel target for future therapies for ARDS and other inflammatory lung diseases.
Collapse
Affiliation(s)
- Christine L. Vigeland
- Department of MedicineUniversity of North CarolinaChapel HillNorth Carolina
- Marsico Lung InstituteUniversity of North CarolinaChapel HillNorth Carolina
| | - Henry S. Beggs
- Department of MedicineUniversity of North CarolinaChapel HillNorth Carolina
- Marsico Lung InstituteUniversity of North CarolinaChapel HillNorth Carolina
| | - Samuel L. Collins
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMaryland
| | - Yee Chan‐Li
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMaryland
| | - Jonathan D. Powell
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMaryland
| | - Claire M. Doerschuk
- Department of MedicineUniversity of North CarolinaChapel HillNorth Carolina
- Marsico Lung InstituteUniversity of North CarolinaChapel HillNorth Carolina
- Center for Airways DiseaseUniversity of North CarolinaChapel HillNorth Carolina
| | - Maureen R. Horton
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMaryland
| |
Collapse
|
20
|
Zhang X, Tan X, Liu Y, You W, Liu G, Liu X, Jin Q, Wei C, Wan F, Zhao H. Alanyl-glutamine ameliorates lipopolysaccharide-induced inflammation and barrier function injury in bovine jejunum epithelial cells. Biochem Cell Biol 2019; 97:670-680. [PMID: 30773024 DOI: 10.1139/bcb-2018-0320] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to investigate the effects of alanyl-glutamine (Ala-Gln) on the regulation of lipopolysaccharide (LPS)-induced inflammation and barrier function in bovine jejunum epithelial cells (BJECs). BJECs were exposed (or not) to 1 μg/mL LPS for 24 h to generate a pro-inflammatory model. The cells were then treated with different concentrations of Ala-Gln (0.25, 0.5, 1.0, 2.0, or 4.0 mmol/L) to detect any regulatory effects on the inflammation and barrier function of BJECs. LPS decreased cell viability and enhanced the production of the pro-inflammatory cytokines interleukin (IL)-6 and IL-8. LPS induced inflammation and damaged the barrier function of BJECs, as evidenced by up-regulated mRNA and protein expression of inflammatory factors and down-regulated expression of tight junction proteins. Conversely, Ala-Gln rescued the decrease in cell viability and prevented the accumulation of ILs after LPS exposure by reducing the mRNA and protein expression levels of inflammatory factors. In addition, Ala-Gln induced the mRNA and protein expression of multiple tight junction proteins, and thus reconstituted the barrier function of BJECs. In conclusion, Ala-Gln attenuates injury from inflammation and repairs damaged intestinal barrier induced with LPS, suggesting its potential as a therapeutic agent against intestinal inflammation in mammals.
Collapse
Affiliation(s)
- Xianglun Zhang
- Institute of Animal Science and Veterinary Medicine, Shandong Key Lab of Animal Disease Control and Breeding, Shandong Provincial Testing Center of Beef Cattle Performance, Shandong Provincial Engineering Technology Center of Animal Healthy Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, People's Republic of China
| | - Xiuwen Tan
- Institute of Animal Science and Veterinary Medicine, Shandong Key Lab of Animal Disease Control and Breeding, Shandong Provincial Testing Center of Beef Cattle Performance, Shandong Provincial Engineering Technology Center of Animal Healthy Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, People's Republic of China
| | - Yifan Liu
- Institute of Animal Science and Veterinary Medicine, Shandong Key Lab of Animal Disease Control and Breeding, Shandong Provincial Testing Center of Beef Cattle Performance, Shandong Provincial Engineering Technology Center of Animal Healthy Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, People's Republic of China
| | - Wei You
- Institute of Animal Science and Veterinary Medicine, Shandong Key Lab of Animal Disease Control and Breeding, Shandong Provincial Testing Center of Beef Cattle Performance, Shandong Provincial Engineering Technology Center of Animal Healthy Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, People's Republic of China
| | - Guifen Liu
- Institute of Animal Science and Veterinary Medicine, Shandong Key Lab of Animal Disease Control and Breeding, Shandong Provincial Testing Center of Beef Cattle Performance, Shandong Provincial Engineering Technology Center of Animal Healthy Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, People's Republic of China
| | - Xiaomu Liu
- Institute of Animal Science and Veterinary Medicine, Shandong Key Lab of Animal Disease Control and Breeding, Shandong Provincial Testing Center of Beef Cattle Performance, Shandong Provincial Engineering Technology Center of Animal Healthy Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, People's Republic of China
| | - Qing Jin
- Institute of Animal Science and Veterinary Medicine, Shandong Key Lab of Animal Disease Control and Breeding, Shandong Provincial Testing Center of Beef Cattle Performance, Shandong Provincial Engineering Technology Center of Animal Healthy Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, People's Republic of China
| | - Chen Wei
- Institute of Animal Science and Veterinary Medicine, Shandong Key Lab of Animal Disease Control and Breeding, Shandong Provincial Testing Center of Beef Cattle Performance, Shandong Provincial Engineering Technology Center of Animal Healthy Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, People's Republic of China
| | - Fachun Wan
- Institute of Animal Science and Veterinary Medicine, Shandong Key Lab of Animal Disease Control and Breeding, Shandong Provincial Testing Center of Beef Cattle Performance, Shandong Provincial Engineering Technology Center of Animal Healthy Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, People's Republic of China.,College of Life Sciences, Shandong Normal University, Jinan 250114, People's Republic of China
| | - Hongbo Zhao
- Institute of Animal Science and Veterinary Medicine, Shandong Key Lab of Animal Disease Control and Breeding, Shandong Provincial Testing Center of Beef Cattle Performance, Shandong Provincial Engineering Technology Center of Animal Healthy Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, People's Republic of China
| |
Collapse
|
21
|
Lei CS, Wu JM, Lee PC, Kuo TC, Chen PD, Hou YC, Yeh SL, Lin MT. Antecedent Administration of Glutamine Benefits the Homeostasis of CD4 + T Cells and Attenuates Lung Injury in Mice With Gut-Derived Polymicrobial Sepsis. JPEN J Parenter Enteral Nutr 2019; 43:927-936. [PMID: 30714626 DOI: 10.1002/jpen.1505] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/03/2019] [Indexed: 01/13/2023]
Abstract
BACKGROUND Sepsis is a syndrome with CD4+ T-cell dysfunction and dysregulation of T helper (Th) and regulatory T (Treg) cells. Glutamine (Gln) is a nutrient with immunomodulatory properties. This study investigated the effects of dietary Gln pretreatment on Th and Treg cell homeostasis and lung injury in mice with gut-derived polymicrobial sepsis. METHODS Mice were randomly assigned to 4 groups with 2 control (C and G) and 2 sepsis groups (SC and SG). The C and SC groups were fed a common semipurified diet, whereas the G and SG groups received an identical diet except that part of the casein was replaced by Gln. Mice were administered these diets for 2 weeks. Then mice in the control groups underwent a sham operation, whereas operations in the sepsis groups were performed with cecal ligation and puncture. Mice were killed 24 hours after the surgery. Blood, spleens, and lungs were collected for further examination. RESULTS Sepsis resulted in a decreased blood T-lymphocyte percentage, whereas percentages of interferon-γ-expressing, interleukin (IL)-4-expressing, and IL-17-expressing CD4+ T cells were upregulated. Compared with the SC group, Gln administration before sepsis reduced blood Th1, Th2, and Th17 but increased Treg percentages. Also, percentages of CD69-expressing CD4+ and CD8+ cells in the spleen increased. Concomitant with the decreased plasma IL-6 and keratinocyte-derived chemokine levels, the SG group exhibited a lower injury score of the lungs. CONCLUSIONS Pretreatment with Gln may elicit more balanced Th polarization, alleviate inflammatory response, and attenuate lung injury induced by polymicrobial sepsis.
Collapse
Affiliation(s)
- Cing-Syuan Lei
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jin-Ming Wu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Chu Lee
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ting-Chun Kuo
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Da Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chen Hou
- Master Program in Food Safety, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Sung-Ling Yeh
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Ming-Tsan Lin
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
22
|
Babajafari S, Akhlaghi M, Mazloomi SM, Ayaz M, Noorafshan A, Jafari P, Hojhabrimanesh A. The effect of isolated soy protein adjunctive with flaxseed oil on markers of inflammation, oxidative stress, acute phase proteins, and wound healing of burn patients; a randomized clinical trial. Burns 2018; 44:140-149. [DOI: 10.1016/j.burns.2017.05.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 10/19/2022]
|
23
|
Dai M, Wang F, Zou Z, Xiao G, Chen H, Yang H. Metabolic regulations of a decoction of Hedyotis diffusa in acute liver injury of mouse models. Chin Med 2017; 12:35. [PMID: 29296119 PMCID: PMC5738817 DOI: 10.1186/s13020-017-0159-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/14/2017] [Indexed: 01/22/2023] Open
Abstract
Background Dysfunctional metabolisms are contributed to LPS/GALN-induced hepatitis. However, whether Hedyotis diffusa (HD) employs metabolic strategies against hepatitis is unknown. Methods We use the cytokines expression, levels of serum alanine transaminase and aspartate transaminase, survival and histological analysis to measure the effect of decoction of HD on acute severe hepatitis of mouse induced by LPS/GALN. Meanwhile, we utilize GC/MS-based metabolomics to characterize the variation of metabolomes. Results The present study shows the relieving liver damage in HD decoction-treated mice. Metabolic category using differential metabolites indicates the lower percentage of carbohydrates in LPS/GALN + HD group than LPS/GALN group, revealing the value of carbohydrate metabolism in HD decoction-administrated mouse liver. Further pathway enrichment analysis proposes that citrate cycle, galactose metabolism, and starch and sucrose metabolism are three important carbohydrate metabolisms that involve in the protective effect of decoction of HD during acute hepatitis. Furthermore, other important enrichment pathways are biosynthesis of unsaturated fatty acids, alanine, aspartate and glutamate metabolism, and arginine and proline metabolism. Fatty acids or amino acids involved in above-mentioned pathways are also detected in high loading distribution on IC01 and IC02, thereby manifesting the significance of these metabolites. Other key metabolites detect in ICA analysis were cholesterol, lactic acid and tryptophan. Conclusions The variation tendency of above-mentioned metabolites is totally consistent with the protective nature of decoction of HD. These findings give a viewpoint that HD decoction-effected metabolic strategies are linked to underlying mechanisms of decoction of HD and highlight the importance of metabolic mechanisms against hepatitis. Electronic supplementary material The online version of this article (10.1186/s13020-017-0159-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min Dai
- Traditional Chinese Medicine Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| | - Fenglin Wang
- Traditional Chinese Medicine Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| | - Zengcheng Zou
- Traditional Chinese Medicine Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| | - Gemin Xiao
- Traditional Chinese Medicine Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| | - Hongjie Chen
- Traditional Chinese Medicine Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| | - Hongzhi Yang
- Traditional Chinese Medicine Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630 China
| |
Collapse
|
24
|
Modulation of mitogen‑activated protein kinase attenuates sepsis‑induced acute lung injury in acute respiratory distress syndrome rats. Mol Med Rep 2017; 16:9652-9658. [PMID: 29039541 DOI: 10.3892/mmr.2017.7811] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 08/10/2017] [Indexed: 11/05/2022] Open
Abstract
Sepsis is the most important predisposing cause inducing acute respiratory distress syndrome (ARDS); however, the mechanism of sepsis leading to the development of ARDS remains to be elucidated. Suppression of the mitogen‑activated protein kinase (MAPK) signal by blocking the phosphorylation of Jun N‑terminal kinase (JNK) and p38 in lung tissues could alleviate acute lung injury induced by sepsis. MAPK signaling may have a crucial role in development of the sepsis‑induced acute lung injury. The specific inhibitors of JNK and p38 MAPK, SP600125 and SB203580, were administrated by intragastric injection 4 h before induction of ARDS after cecal ligation and puncture (CLP). Rats were sacrificed at 1, 6 or 24 h after CLP challenge. The histological evaluation, lung water content, and biochemical analysis were performed. The results revealed that the JNK and p38 MAPK inhibitor improved lung permeability, attenuated system inflammation, further alleviated the lung injury induced by sepsis. In conclusion, JNK and p38 MAPK signaling are essential for the development of ARDS following sepsis. Further studies are needed to illuminate the detailed mechanisms of JNK and p38 MAPK signaling in sepsis‑induced ARDS.
Collapse
|
25
|
Wang H, Dong Y, Cai Y. Alanyl-glutamine prophylactically protects against lipopolysaccharide-induced acute lung injury by enhancing the expression of HSP70. Mol Med Rep 2017; 16:2807-2813. [DOI: 10.3892/mmr.2017.6896] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 03/02/2017] [Indexed: 11/06/2022] Open
|
26
|
Aqeel M, Ahmad S, Patel JJ, Rice TW. Immunonutrition in Acute Respiratory Distress Syndrome. CURRENT PULMONOLOGY REPORTS 2017. [DOI: 10.1007/s13665-017-0171-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
27
|
Gomes PC, Caporossi C, Aguilar-Nascimento JE, Silva AMCD, Araujo VMTD. Residual gastric volume evaluation with ultrasonography after ingestion of carbohydrate- or carbohydrate plus glutamine-enriched beverages: a randomized, crossover clinical trial with healthy volunteers. ARQUIVOS DE GASTROENTEROLOGIA 2017; 54:33-36. [PMID: 28079236 DOI: 10.1590/s0004-2803.2017v54n1-06] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/22/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND - Abbreviation of preoperative fasting to 2 hours with maltodextrin (CHO)-enriched beverage is a safe procedure and may enhance postoperative recovery. Addition of glutamine (GLN) to CHO beverages may include potential benefits to the metabolism. However, by adding a nitrogenous source to CHO beverages, gastric emptying may be delayed and increase the risk of bronchoaspiration during anesthesia. OBJECTIVE - In this study of safety, we aimed at investigating the residual gastric volume (RGV) 2 hours after the intake of either CHO beverage alone or CHO beverage combined with GLN. METHODS - We performed a randomized, crossover clinical trial. We assessed RGV by means of abdominal ultrasonography (US) in 20 healthy volunteers (10 males and 10 females) after an overnight fast of 8 hours. Then, they were randomized to receive 600 mL (400 mL immediately after US followed by another 200 mL 2 hours afterwards) of either CHO (12.5% maltodextrin) or CHO-GLN (12.5% maltodextrin plus 15 g GLN). Two sequential US evaluations were done at 120 and 180 minutes after ingestion of the second dose. The interval of time between ingestion of the two types of beverages was 2 weeks. RESULTS - The mean (SD) RGV observed after 8 hours fasting (13.56±13.25 mL) did not statistically differ (P>0.05) from the RGV observed after ingesting CHO beverage at both 120 (16.32±11.78 mL) and 180 minutes (14.60±10.39 mL). The RGV obtained at 120 (15.63±18.83 mL) and 180 (13.65±10.27 mL) minutes after CHO-GLN beverage also was not significantly different from the fasting condition. CONCLUSION - The RGV at 120 and 180 minutes after ingestion of CHO beverage combined with GLN is similar to that observed after an overnight fast.
Collapse
Affiliation(s)
- Paulo Cesar Gomes
- Faculdade de Medicina, Universidade Federal de Mato Grosso, MT, Brasil
| | | | | | | | | |
Collapse
|
28
|
Leite JSM, Cruzat VF, Krause M, Homem de Bittencourt PI. Physiological regulation of the heat shock response by glutamine: implications for chronic low-grade inflammatory diseases in age-related conditions. ACTA ACUST UNITED AC 2016. [DOI: 10.1186/s41110-016-0021-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
29
|
Kratochwill K, Boehm M, Herzog R, Gruber K, Lichtenauer AM, Kuster L, Csaicsich D, Gleiss A, Alper SL, Aufricht C, Vychytil A. Addition of Alanyl-Glutamine to Dialysis Fluid Restores Peritoneal Cellular Stress Responses - A First-In-Man Trial. PLoS One 2016; 11:e0165045. [PMID: 27768727 PMCID: PMC5074513 DOI: 10.1371/journal.pone.0165045] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/01/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Peritonitis and ultrafiltration failure remain serious complications of chronic peritoneal dialysis (PD). Dysfunctional cellular stress responses aggravate peritoneal injury associated with PD fluid exposure, potentially due to peritoneal glutamine depletion. In this randomized cross-over phase I/II trial we investigated cytoprotective effects of alanyl-glutamine (AlaGln) addition to glucose-based PDF. METHODS In a prospective randomized cross-over design, 20 stable PD outpatients underwent paired peritoneal equilibration tests 4 weeks apart, using conventional acidic, single chamber 3.86% glucose PD fluid, with and without 8 mM supplemental AlaGln. Heat-shock protein 72 expression was assessed in peritoneal effluent cells as surrogate parameter of cellular stress responses, complemented by metabolomics and functional immunocompetence assays. RESULTS AlaGln restored peritoneal glutamine levels and increased the primary outcome heat-shock protein expression (effect 1.51-fold, CI 1.07-2.14; p = 0.022), without changes in peritoneal ultrafiltration, small solute transport, or biomarkers reflecting cell mass and inflammation. Further effects were glutamine-like metabolomic changes and increased ex-vivo LPS-stimulated cytokine release from healthy donor peripheral blood monocytes. In patients with a history of peritonitis (5 of 20), AlaGln supplementation decreased dialysate interleukin-8 levels. Supplemented PD fluid also attenuated inflammation and enhanced stimulated cytokine release in a mouse model of PD-associated peritonitis. CONCLUSION We conclude that AlaGln-supplemented, glucose-based PD fluid can restore peritoneal cellular stress responses with attenuation of sterile inflammation, and may improve peritoneal host-defense in the setting of PD.
Collapse
Affiliation(s)
- Klaus Kratochwill
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Zytoprotec GmbH, Vienna, Austria
| | - Michael Boehm
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Rebecca Herzog
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Zytoprotec GmbH, Vienna, Austria
| | - Katharina Gruber
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Anton Michael Lichtenauer
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Zytoprotec GmbH, Vienna, Austria
| | - Lilian Kuster
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Zytoprotec GmbH, Vienna, Austria
| | - Dagmar Csaicsich
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Andreas Gleiss
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Seth L. Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Christoph Aufricht
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Andreas Vychytil
- Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
30
|
Determination of the anti-inflammatory and cytoprotective effects of l-glutamine and l-alanine, or dipeptide, supplementation in rats submitted to resistance exercise. Br J Nutr 2016; 116:470-9. [PMID: 27215379 DOI: 10.1017/s0007114516001999] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We evaluated the effects of chronic oral supplementation with l-glutamine and l-alanine in their free form or as the dipeptide l-alanyl-l-glutamine (DIP) on muscle damage, inflammation and cytoprotection, in rats submitted to progressive resistance exercise (RE). Wistar rats (n 8/group) were submitted to 8-week RE, which consisted of climbing a ladder with progressive loads. In the final 21 d before euthanasia, supplements were delivered in a 4 % solution in drinking water. Glutamine, creatine kinase (CK), lactate dehydrogenase (LDH), TNF-α, specific IL (IL-1β, IL-6 and IL-10) and monocyte chemoattractant protein-1 (MCP-1) levels were evaluated in plasma. The concentrations of glutamine, TNF-α, IL-6 and IL-10, as well as NF-κB activation, were determined in extensor digitorum longus (EDL) skeletal muscle. HSP70 level was assayed in EDL and peripheral blood mononuclear cells (PBMC). RE reduced glutamine concentration in plasma and EDL (P<0·05 v. sedentary group). However, l-glutamine supplements (l-alanine plus l-glutamine (GLN+ALA) and DIP groups) restored glutamine levels in plasma (by 40 and 58 %, respectively) and muscle (by 93 and 105 %, respectively). GLN+ALA and DIP groups also exhibited increased level of HSP70 in EDL and PBMC, consistent with the reduction of NF-κB p65 activation and cytokines in EDL. Muscle protection was also indicated by attenuation in plasma levels of CK, LDH, TNF-α and IL-1β, as well as an increase in IL-6, IL-10 and MCP-1. Our study demonstrates that chronic oral l-glutamine treatment (given with l-alanine or as dipeptide) following progressive RE induces cyprotective effects mediated by HSP70-associated responses to muscle damage and inflammation.
Collapse
|
31
|
Girven M, Dugdale HF, Owens DJ, Hughes DC, Stewart CE, Sharples AP. l-glutamine Improves Skeletal Muscle Cell Differentiation and Prevents Myotube Atrophy After Cytokine (TNF-α) Stress Via Reduced p38 MAPK Signal Transduction. J Cell Physiol 2016; 231:2720-32. [PMID: 26991744 DOI: 10.1002/jcp.25380] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/15/2016] [Indexed: 02/06/2023]
Abstract
Tumour Necrosis Factor-Alpha (TNF-α) is chronically elevated in conditions where skeletal muscle loss occurs. As l-glutamine can dampen the effects of inflamed environments, we investigated the role of l-glutamine in both differentiating C2C12 myoblasts and existing myotubes in the absence/presence of TNF-α (20 ng · ml(-1) ) ± l-glutamine (20 mM). TNF-α reduced the proportion of cells in G1 phase, as well as biochemical (CK activity) and morphological differentiation (myotube number), with corresponding reductions in transcript expression of: Myogenin, Igf-I, and Igfbp5. Furthermore, when administered to mature myotubes, TNF-α induced myotube loss and atrophy underpinned by reductions in Myogenin, Igf-I, Igfbp2, and glutamine synthetase and parallel increases in Fox03, Cfos, p53, and Bid gene expression. Investigation of signaling activity suggested that Akt and ERK1/2 were unchanged, JNK increased (non-significantly) whereas P38 MAPK substantially and significantly increased in both myoblasts and myotubes in the presence of TNF-α. Importantly, 20 mM l-glutamine reduced p38 MAPK activity in TNF-α conditions back to control levels, with a corresponding rescue of myoblast differentiation and a reversal of atrophy in myotubes. l-glutamine resulted in upregulation of genes associated with growth and survival including; Myogenin, Igf-Ir, Myhc2 & 7, Tnfsfr1b, Adra1d, and restored atrophic gene expression of Fox03 back to baseline in TNF-α conditions. In conclusion, l-glutamine supplementation rescued suppressed muscle cell differentiation and prevented myotube atrophy in an inflamed environment via regulation of p38 MAPK. l-glutamine administration could represent an important therapeutic strategy for reducing muscle loss in catabolic diseases and inflamed ageing. J. Cell. Physiol. 9999: 231: 2720-2732, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Matthew Girven
- Stem Cells, Ageing and Molecular Physiology Research Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
| | - Hannah F Dugdale
- Stem Cells, Ageing and Molecular Physiology Research Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
| | - Daniel J Owens
- Stem Cells, Ageing and Molecular Physiology Research Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom.,Sorbonne Universités, UPMC University of Paris 06, INSERM UMRS974, CNRS FRE3617, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, Paris 13, France
| | - David C Hughes
- Stem Cells, Ageing and Molecular Physiology Research Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom.,Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California
| | - Claire E Stewart
- Stem Cells, Ageing and Molecular Physiology Research Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
| | - Adam P Sharples
- Stem Cells, Ageing and Molecular Physiology Research Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
32
|
Bourgoin-Voillard S, Goron A, Seve M, Moinard C. Regulation of the proteome by amino acids. Proteomics 2016; 16:831-46. [DOI: 10.1002/pmic.201500347] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/30/2015] [Accepted: 01/12/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Sandrine Bourgoin-Voillard
- Plateforme de Protéomique PROMETHEE; IAB; University Grenoble Alpes; Grenoble France
- Plateforme de Protéomique PROMETHEE, Institut de Biologie et de Pathologie; CHU de Grenoble; Grenoble France
- Plateforme de Protéomique PROMETHEE; IAB; INSERM; Grenoble France
| | - Arthur Goron
- Laboratory of Fundamental and Applied Bioenergetics (LBFA); University Grenoble Alpes; Grenoble France
- Laboratory of Fundamental and Applied Bioenergetics (LBFA); INSERM; Grenoble France
| | - Michel Seve
- Plateforme de Protéomique PROMETHEE; IAB; University Grenoble Alpes; Grenoble France
- Plateforme de Protéomique PROMETHEE, Institut de Biologie et de Pathologie; CHU de Grenoble; Grenoble France
- Plateforme de Protéomique PROMETHEE; IAB; INSERM; Grenoble France
| | - Christophe Moinard
- Laboratory of Fundamental and Applied Bioenergetics (LBFA); University Grenoble Alpes; Grenoble France
- Laboratory of Fundamental and Applied Bioenergetics (LBFA); INSERM; Grenoble France
| |
Collapse
|
33
|
Geamanu A, Gupta SV, Bauerfeld C, Samavati L. Metabolomics connects aberrant bioenergetic, transmethylation, and gut microbiota in sarcoidosis. Metabolomics 2016; 12:35. [PMID: 27489531 PMCID: PMC4960975 DOI: 10.1007/s11306-015-0932-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Sarcoidosis is a systemic granulomatous disease of unknown etiology. Granulomatous inflammation in sarcoidosis may affect multiple organs, including the lungs, skin, CNS, and the eyes, leading to severe morbidity and mortality. The underlying mechanisms for sustained inflammation in sarcoidosis are unknown. We hypothesized that metabolic changes play a critical role in perpetuation of inflammation in sarcoidosis. 1H nuclear magnetic resonance (NMR)-based untargeted metabolomic analysis was used to identify circulating molecules in serum to discriminate sarcoidosis patients from healthy controls. Principal component analyses (PCA) were performed to identify different metabolic markers and explore the changes of associated biochemical pathways. Using Chenomx 7.6 NMR Suite software, we identified and quantified metabolites responsible for such separation in the PCA models. Quantitative analysis showed that the levels of metabolites, such as 3-hydroxybutyrate, acetoacetate, carnitine, cystine, homocysteine, pyruvate, and trimethylamine N-oxide were significantly increased in sarcoidosis patients. Interestingly, succinate, a major intermediate metabolite involved in the tricyclic acid cycle was significantly decreased in sarcoidosis patients. Application of integrative pathway analyses identified deregulation of butanoate, ketone bodies, citric cycle metabolisms, and transmethylation. This may be used for development of new drugs or nutritional modification.
Collapse
Affiliation(s)
- Andreea Geamanu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Wayne State University School of Medicine and Detroit Medical Center, 3990 John R., 3 Hudson, Detroit, MI 48201, USA
| | - Smiti V. Gupta
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48201, USA
| | - Christian Bauerfeld
- Division of Pediatric Critical Care, Department of Pediatrics, Children’s Hospital of Michigan, Detroit, MI 48201, USA
| | - Lobelia Samavati
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Wayne State University School of Medicine and Detroit Medical Center, 3990 John R., 3 Hudson, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
34
|
Innate immunity gene expression changes in critically ill patients with sepsis and disease-related malnutrition. Cent Eur J Immunol 2015; 40:311-24. [PMID: 26648775 PMCID: PMC4655381 DOI: 10.5114/ceji.2015.54593] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 07/14/2015] [Indexed: 12/26/2022] Open
Abstract
The aim of this study was an attempt to determine whether the expression of genes involved in innate antibacterial response (TL R2, NOD 1, TRAF6, HMGB 1 and Hsp70) in peripheral blood leukocytes in critically ill patients, may undergo significant changes depending on the severity of the infection and the degree of malnutrition. The study was performed in a group of 128 patients with infections treated in the intensive care and surgical ward. In 103/80.5% of patients, infections had a severe course (sepsis, severe sepsis, septic shock, mechanical ventilation of the lungs). Clinical monitoring included diagnosis of severe infection (according to the criteria of the ACC P/SCC M), assessment of severity of the patient condition and risk of death (APACHE II and SAPS II), nutritional assessment (NRS 2002 and SGA scales) and the observation of the early results of treatment. Gene expression at the mRNA level was analyzed by real-time PCR. The results of the present study indicate that in critically ill patients treated in the IC U there are significant disturbances in the expression of genes associated with innate antimicrobial immunity, which may have a significant impact on the clinical outcome. The expression of these genes varies depending on the severity of the patient condition, severity of infection and nutritional status. Expression disorders of genes belonging to innate antimicrobial immunity should be diagnosed as early as possible, monitored during the treatment and taken into account during early therapeutic treatment (including early nutrition to support the functions of immune cells).
Collapse
|
35
|
Glutamine may repress the weak LPS and enhance the strong heat shock induction of monocyte and lymphocyte HSP72 proteins but may not modulate the HSP72 mRNA in patients with sepsis or trauma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:806042. [PMID: 26550577 PMCID: PMC4621332 DOI: 10.1155/2015/806042] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/23/2015] [Accepted: 08/31/2015] [Indexed: 12/29/2022]
Abstract
Objective. We assessed the lipopolysaccharide (LPS) or heat shock (HS) induction of heat shock protein-72 (HSP72) in peripheral blood mononuclear cells (PBMCs) of patients with severe sepsis (SS) or trauma-related systemic inflammatory response syndrome (SIRS), compared to healthy individuals (H); we also investigated any pre- or posttreatment modulating glutamine (Gln) effect. Methods. SS (11), SIRS (10), and H (19) PBMCs were incubated with 1 μg/mL LPS or 43°HS. Gln 10 mM was either added 1 h before or 1 h after induction or was not added at all. We measured monocyte (m), lymphocyte (l), mRNA HSP72, HSP72 polymorphisms, interleukins (ILs), monocyte chemoattractant protein-1 (MCP-1), and cortisol levels. Results. Baseline lHSP72 was higher in SS (p < 0.03), and mHSP72 in SIRS (p < 0.02), compared to H. Only HS induced l/mHSP72/mRNA HSP72; LPS induced IL-6, IL-8, IL-10, and MCP-1. Induced mRNA was related to l/mHSP72, and was related negatively to cytokines. Intracellular l/mHSP72/HSP72 mRNA was related to serum ILs, not being influenced by cortisol, illness severity, and HSP72 polymorphisms. Gln did not induce mRNA in any group but modified l/mHSP72 after LPS/HS induction unpredictably. Conclusions. HSP72 mRNA and l/mHSP72 are higher among critically ill patients, further induced by HS, not by LPS. HSP72 proteins and HSP72 mRNA are related to serum ILs and are negatively related to supernatant cytokines, not being influenced by HSP72 polymorphisms, cortisol, or illness severity. Gln may depress l/mHSP72 after LPS exposure and enhance them after HS induction, but it may not affect early induced HSP72 mRNA.
Collapse
|
36
|
Marino LV, Pathan N, Meyer RW, Wright VJ, Habibi P. An in vitro model to consider the effect of 2 mM glutamine and KNK437 on endotoxin-stimulated release of heat shock protein 70 and inflammatory mediators. Nutrition 2015; 32:375-83. [PMID: 26706024 DOI: 10.1016/j.nut.2015.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/13/2015] [Accepted: 09/13/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Glutamine has been shown to promote the release of heat shock protein 70 (HSP70) both within experimental in vitro models of sepsis and in adults with septic shock. This study aimed to investigate the effects of 2 mM glutamine and an inhibitor of HSP70 (KNK437) on the release of HSP70 and inflammatory mediators in healthy adult volunteers. METHODS An in vitro whole blood endotoxin stimulation assay was used. RESULTS The addition of 2 mM glutamine significantly increased HSP70 levels over time (P < 0.05). HSP70 release had a positive correlation at 4 h with IL-1 β (r = 0.51, P = 0.03) and an inverse correlation with TNF-α (r = -0.56, P = 0.02) and IL-8 levels (r = -0.52, P = 0.03), and there were no significant correlations between HSP70 and IL6 or IL-10 or glutamine. Glutamine supplementation significantly (P < 0.05) attenuated the release of IL-10 at 4 h and IL-8 at 24 h, compared with conditions without glutamine. In endotoxin-stimulated blood there were no significant differences in the release of IL-6, TNF-α, and IL-1 β with glutamine supplementation at 4 and 24 h. However, glutamine supplementation (2 mM) appeared to attenuate the release of inflammatory mediators (IL-1 β, IL-6, TNF-α), although this effect was not statistically significant. The addition of KNK437, a HSP70 inhibitor, significantly diminished HSP70 release, which resulted in lower levels of inflammatory mediators (P < 0.05). CONCLUSION Glutamine supplementation promotes HSP70 release in an experimental model of sepsis. After the addition of KNK437, the effects of glutamine on HSP70 and inflammatory mediator release appear to be lost, suggesting that HSP70 in part orchestrates the inflammatory mediator response to sepsis. The clinical implications require further investigation.
Collapse
Affiliation(s)
- Luise V Marino
- Department of Paediatrics, Imperial College London, London, United Kingdom.
| | - Nazima Pathan
- Department of Paediatrics, School of Clinical Medicine, Cambridge University, Cambridge, United Kingdom
| | - Rosan W Meyer
- Department of Gastroenterology, Great Ormond Street Hospital for Sick Children, London, United Kingdom
| | - Victoria J Wright
- Department of Paediatrics, Imperial College London, London, United Kingdom
| | - Parviz Habibi
- Department of Paediatrics, Imperial College London, London, United Kingdom
| |
Collapse
|
37
|
Effects of enteral nutrition with parenteral glutamine supplementation on the immunological function in septic rats. Br J Nutr 2015; 113:1712-22. [PMID: 26067806 DOI: 10.1017/s0007114515001099] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The aim of the present study was to investigate the effects of enteral nutrition (EN) with parenteral glutamine (GLN) supplementation on inflammatory response, lymphatic organ apoptosis, immunological function and survival in septic rats by caecal ligation and puncture (CLP). Male rats were randomly assigned into two experimental groups and two sham CLP control groups (n 10 per group). After CLP or sham CLP model and nutrition programme were completed, the GLN concentrations of plasma and tissues and several indices of immunological function including serum Ig content, circulating lymphocyte number, the CD4:CD8 ratio, the neutrophil phagocytosis index (NPI), the organ index and apoptosis of thymus and spleen, and plasma cytokine levels were determined. Moreover, the survival in septic rats was observed. The results revealed that EN with parenteral GLN supplementation remarkably increased the GLN concentrations of plasma and tissues, serum Ig content, the circulating lymphocyte number, the CD4:CD8 ratio, the indexes of thymus and spleen, NPI and survival compared with the control group (P< 0·05). In contrast, the apoptosis of thymus and spleen and the levels of TNF-α, IL-1β and IL-6 in plasma were obviously decreased compared with the control group (P< 0·05). These results show that EN with parenteral GLN supplementation diminished the release of inflammatory cytokines, attenuated lymphatic organ apoptosis, enhanced the immunological function and improved survival in septic rats.
Collapse
|
38
|
Chen CM, Cheng KC, Li CF, Zhang H. The protective effects of glutamine in a rat model of ventilator-induced lung injury. J Thorac Dis 2015; 6:1704-13. [PMID: 25589963 DOI: 10.3978/j.issn.2072-1439.2014.11.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 09/24/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND The mortality rate of patients with acute respiratory distress syndrome (ARDS) is still high despite the use of protective ventilatory strategies. We sought to examine the pharmacological effects of glutamine (GLN) in a two-hit model of endotoxin-induced inflammation followed by ventilator-induced lung injury (VILI). We hypothesized that the administration of GLN ameliorates the VILI. METHODS Sprague-Dawley rats were anesthetized and given lipopolysaccharide (LPS) intratracheally as a first hit to induce lung inflammation, followed 24 h later by a second hit of mechanical ventilation (MV) with either low tidal volume (6 mL/kg) with 5 cmH2O of positive end-expiratory pressure (PEEP) or high tidal volume (22 mL/kg) with zero PEEP for 4 h. GLN or lactated Ringer's solution as the placebo was administered intravenously 15 min prior to MV. RESULTS In the LPS-challenged rats ventilated with high tidal volume, the treatment with GLN improved lung injury indices, lung mechanics and cytokine responses compared with the placebo group. CONCLUSIONS The administration of GLN given immediately prior to MV may be beneficial in the context of reducing VILI.
Collapse
Affiliation(s)
- Chin-Ming Chen
- 1 Department of Recreation and Healthcare Management, Chia Nan University of Pharmacy & Science, Tainan, Taiwan ; 2 Department of Critical Care Medicine, Chi-Mei Medical Center and Chang Jung Christian University, Tainan, Taiwan ; 3 Section of Respiratory Care, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan ; 4 Department of Safety Health and Environment Engineering, Chung Hwa University of Medical Technology, Tainan, Taiwan ; 5 Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan ; 6 Keenan Research Center for Biomedical Science of St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Kuo-Chen Cheng
- 1 Department of Recreation and Healthcare Management, Chia Nan University of Pharmacy & Science, Tainan, Taiwan ; 2 Department of Critical Care Medicine, Chi-Mei Medical Center and Chang Jung Christian University, Tainan, Taiwan ; 3 Section of Respiratory Care, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan ; 4 Department of Safety Health and Environment Engineering, Chung Hwa University of Medical Technology, Tainan, Taiwan ; 5 Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan ; 6 Keenan Research Center for Biomedical Science of St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Chien-Feng Li
- 1 Department of Recreation and Healthcare Management, Chia Nan University of Pharmacy & Science, Tainan, Taiwan ; 2 Department of Critical Care Medicine, Chi-Mei Medical Center and Chang Jung Christian University, Tainan, Taiwan ; 3 Section of Respiratory Care, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan ; 4 Department of Safety Health and Environment Engineering, Chung Hwa University of Medical Technology, Tainan, Taiwan ; 5 Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan ; 6 Keenan Research Center for Biomedical Science of St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Haibo Zhang
- 1 Department of Recreation and Healthcare Management, Chia Nan University of Pharmacy & Science, Tainan, Taiwan ; 2 Department of Critical Care Medicine, Chi-Mei Medical Center and Chang Jung Christian University, Tainan, Taiwan ; 3 Section of Respiratory Care, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan ; 4 Department of Safety Health and Environment Engineering, Chung Hwa University of Medical Technology, Tainan, Taiwan ; 5 Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan ; 6 Keenan Research Center for Biomedical Science of St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
39
|
Marino LV, Pathan N, Meyer R, Wright VJ, Habibi P. The effect of 2 mMol glutamine supplementation on HSP70 and TNF-α release by LPS stimulated blood from healthy children. Clin Nutr 2014; 34:1195-201. [PMID: 25556350 DOI: 10.1016/j.clnu.2014.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 12/14/2014] [Accepted: 12/16/2014] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Glutamine has been shown to promote heat shock protein 70 (HSP70) release both within experimental in vitro models of sepsis (2-10 mM) and in adults post trauma (0.5 g/kg), although the efficacy varies and is dependent on the model used. The effect of glutamine supplementation on HSP70 release in children is less clear. Therefore, the aim of this study was to investigate the effect of 2 mM glutamine added to incubation media on HSP70 and inflammatory mediator release in an in vitro model of paediatric sepsis using whole blood from healthy paediatric volunteers. METHODS An in vitro whole blood endotoxin stimulation model using 1 μg/ml lipopolysaccharide (LPS) over a 24 h time period was used to investigate the effects of 2 mM glutamine on HSP70 and inflammatory mediator release in healthy children. RESULTS The addition of 2 mM glutamine to the incubation media significantly increased HSP70 release over time (p < 0.05). This was associated with an early pro-inflammatory effect on TNF-α release at 4 h (p < 0.005) which was not seen at 24 h. There was a non significant trend towards higher levels of IL-6 and IL-10 following the addition of 2 mM glutamine, which appears to differ from the response reported in adult and animal models. CONCLUSION Glutamine supplementation of incubation media promotes HSP70 and early TNF- α release in an in vitro model using blood samples from healthy children.
Collapse
Affiliation(s)
- L V Marino
- Department of Paediatrics, Imperial College, London, UK.
| | - N Pathan
- Department of Paediatrics, School of Clinical Medicine, Cambridge University, UK
| | - R Meyer
- Department of Gastroenterology, Great Ormond Street Hospital for Sick Children, London, UK
| | - V J Wright
- Department of Paediatrics, Imperial College, London, UK
| | - P Habibi
- Department of Paediatrics, Imperial College, London, UK
| |
Collapse
|
40
|
Dietary glutamine prevents the loss of intestinal barrier function and attenuates the increase in core body temperature induced by acute heat exposure. Br J Nutr 2014; 112:1601-10. [PMID: 25322775 DOI: 10.1017/s0007114514002608] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dietary glutamine (Gln) supplementation improves intestinal function in several stressful conditions. Therefore, in the present study, the effects of dietary Gln supplementation on the core body temperature (T core), bacterial translocation (BT) and intestinal permeability of mice subjected to acute heat stress were evaluated. Male Swiss mice (4 weeks old) were implanted with an abdominal temperature sensor and randomly assigned to one of the following groups fed isoenergetic and isoproteic diets for 7 d before the experimental trials: group fed the standard AIN-93G diet and exposed to a high ambient temperature (39°C) for 2 h (H-NS); group fed the AIN-93G diet supplemented with l-Gln and exposed to a high temperature (H-Gln); group fed the standard AIN-93G diet and not exposed to a high temperature (control, C-NS). Mice were orally administered diethylenetriaminepentaacetic acid radiolabelled with technetium (99mTc) for the assessment of intestinal permeability or 99mTc-Escherichia coli for the assessment of BT. Heat exposure increased T core (approximately 41°C during the experimental trial), intestinal permeability and BT to the blood and liver (3 h after the experimental trial) in mice from the H-NS group relative to those from the C-NS group. Dietary Gln supplementation attenuated hyperthermia and prevented the increases in intestinal permeability and BT induced by heat exposure. No correlations were observed between the improvements in gastrointestinal function and the attenuation of hyperthermia by Gln. Our findings indicate that dietary Gln supplementation preserved the integrity of the intestinal barrier and reduced the severity of hyperthermia during heat exposure. The findings also indicate that these Gln-mediated effects occurred through independent mechanisms.
Collapse
|
41
|
Lai CC, Liu WL, Chen CM. Glutamine attenuates acute lung injury caused by acid aspiration. Nutrients 2014; 6:3101-16. [PMID: 25100435 PMCID: PMC4145297 DOI: 10.3390/nu6083101] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/14/2014] [Accepted: 07/24/2014] [Indexed: 01/11/2023] Open
Abstract
Inadequate ventilator settings may cause overwhelming inflammatory responses associated with ventilator-induced lung injury (VILI) in patients with acute respiratory distress syndrome (ARDS). Here, we examined potential benefits of glutamine (GLN) on a two-hit model for VILI after acid aspiration-induced lung injury in rats. Rats were intratracheally challenged with hydrochloric acid as a first hit to induce lung inflammation, then randomly received intravenous GLN or lactated Ringer's solution (vehicle control) thirty min before different ventilator strategies. Rats were then randomized to receive mechanical ventilation as a second hit with a high tidal volume (TV) of 15 mL/kg and zero positive end-expiratory pressure (PEEP) or a low TV of 6 mL/kg with PEEP of 5 cm H2O. We evaluated lung oxygenation, inflammation, mechanics, and histology. After ventilator use for 4 h, high TV resulted in greater lung injury physiologic and biologic indices. Compared with vehicle treated rats, GLN administration attenuated lung injury, with improved oxygenation and static compliance, and decreased respiratory elastance, lung edema, extended lung destruction (lung injury scores and lung histology), neutrophil recruitment in the lung, and cytokine production. Thus, GLN administration improved the physiologic and biologic profiles of this experimental model of VILI based on the two-hit theory.
Collapse
Affiliation(s)
- Chih-Cheng Lai
- Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying Dist., Tainan 73657 Taiwan.
| | - Wei-Lun Liu
- Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying Dist., Tainan 73657 Taiwan.
| | - Chin-Ming Chen
- Department of Recreation and Health-Care Management, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan.
| |
Collapse
|
42
|
Briassouli E, Goukos D, Daikos G, Apostolou K, Routsi C, Nanas S, Briassoulis G. Glutamine suppresses Hsp72 not Hsp90α and is not inducing Th1, Th2, or Th17 cytokine responses in human septic PBMCs. Nutrition 2014; 30:1185-94. [PMID: 24976418 DOI: 10.1016/j.nut.2014.01.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 01/16/2014] [Accepted: 01/30/2014] [Indexed: 11/16/2022]
Abstract
OBJECTIVE L-Alanyl-glutamine (L-Ala-Gln) is a pharmaco-nutrient commonly used in nutrition regimens due to its immunomodulatory effects. In critically ill patients who are septic, L-Ala-Gln was associated with an increase in mortality. The aim of this study was to investigate whether L-Ala-Gln modulated heat shock protein (Hsp)-72, 90-α, T helper (Th)1, Th2, and Th17 cytokine expression in the peripheral blood mononuclear cells (PBMC) of patients with severe sepsis. METHODS Time-dose effects of L-Ala-Gln were compared with those of L-glutamine (L-Gln) and lipopolysaccharide (LPS) and to healthy controls. PBMCs were incubated with 1 or 10 μg/mL LPS, 5 or 10 mM L-Gln, and 5 or 10 mM L-Ala-Gln for different periods of time (0; 4; 24 h) when culture supernatants were harvested. RESULTS In both groups, basal Hsp72 increased over time (P < 0.02); Hsp90-α levels declined in controls (P < 0.02) but remained increased in septic patients (P < 0.02), not exhibiting any significant time-response trend. Both Glns suppressed Hsp72 in septic and controls at 10 mM by 4 h (P < 0.045) and Hsp90-α in the control group by 24 h (P < 0.045). LPS did not induce Hsps in either group. L-Ala-Gln did not induce any of the Th1, Th2, and Th17 cytokines in either group. CONCLUSION High doses of L-Gln or L-Ala-Gln do not induce any of the Th1, Th2, and Th17 cytokines in either healthy or septic human PBMCs. High Gln doses suppress Hsp72 in septic and control PBMCs. Hsp90-α time-series expression declines, contrasting the increasing trend of Hsp72 in healthy controls. Hsp90-α sustains increased levels in septic supernatants, showing a characteristic longitudinal behavior needed further elucidation.
Collapse
Affiliation(s)
- Efrossini Briassouli
- First Department of Internal Medicine, Medical School, University of Athens, Laikon Hospital, Athens, Greece
| | - Dimitris Goukos
- First Department of Internal Medicine, Medical School, University of Athens, Laikon Hospital, Athens, Greece
| | - George Daikos
- First Department of Internal Medicine, Medical School, University of Athens, Laikon Hospital, Athens, Greece
| | - Kleovoulos Apostolou
- First Department of Critical Care, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece
| | - Christina Routsi
- First Department of Critical Care, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece
| | - Serafim Nanas
- First Department of Critical Care, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece
| | - George Briassoulis
- Pediatric Intensive Care Unit, University Hospital, University of Crete, Heraklion, Greece.
| |
Collapse
|
43
|
Heat shock protein 72 expressing stress in sepsis: unbridgeable gap between animal and human studies--a hypothetical "comparative" study. BIOMED RESEARCH INTERNATIONAL 2014; 2014:101023. [PMID: 24524071 PMCID: PMC3912989 DOI: 10.1155/2014/101023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 10/05/2013] [Indexed: 01/30/2023]
Abstract
Heat shock protein 72 (Hsp72) exhibits a protective role during times of increased risk of pathogenic challenge and/or tissue damage. The aim of the study was to ascertain Hsp72 protective effect differences between animal and human studies in sepsis using a hypothetical “comparative study” model.
Forty-one in vivo (56.1%), in vitro (17.1%), or combined (26.8%) animal and 14 in vivo (2) or in vitro (12) human Hsp72 studies (P < 0.0001) were enrolled in the analysis. Of the 14 human studies, 50% showed a protective Hsp72 effect compared to 95.8% protection shown in septic animal studies (P < 0.0001). Only human studies reported Hsp72-associated mortality (21.4%) or infection (7.1%) or reported results (14.3%) to be nonprotective (P < 0.001). In animal models, any Hsp72 induction method tried increased intracellular Hsp72 (100%), compared to 57.1% of human studies (P < 0.02), reduced proinflammatory cytokines (28/29), and enhanced survival (18/18). Animal studies show a clear Hsp72 protective effect in sepsis. Human studies are inconclusive, showing either protection or a possible relation to mortality and infections. This might be due to the fact that using evermore purified target cell populations in animal models, a lot of clinical information regarding the net response that occurs in sepsis is missing.
Collapse
|
44
|
Lin Z, Cai F, Lin N, Ye J, Zheng Q, Ding G. Effects of glutamine on oxidative stress and nuclear factor-κB expression in the livers of rats with nonalcoholic fatty liver disease. Exp Ther Med 2013; 7:365-370. [PMID: 24396406 PMCID: PMC3881322 DOI: 10.3892/etm.2013.1434] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 10/18/2013] [Indexed: 12/22/2022] Open
Abstract
The aim of this study was to investigate the effects of glutamine on the histomorphology of the liver, oxidative stress and nuclear factor-κB (NF-κB) expression in the development of nonalcoholic fatty liver disease (NAFLD). NAFLD was induced in rats by a high-fat diet, and rats in the treatment group were subjected to oral administration of glutamine (1 g/kg/day). Rats from the treatment, model and normal control groups were assessed after 8 and 12 weeks (n=6 per group at each time-point). The levels of glutathione (GSH), malondialdehyde (MDA) and tumor necrosis factor-α (TNF-α) in the liver, and the liver histopathology and NF-κB protein 65 (p65) expression in the liver were assessed. Compared with the control group under the same experimental period, the MDA and TNF-α levels in the liver, the hepatic steatosis and the hepatic expression of NF-κB p65 were significantly higher in the model and the treatment groups (P<0.05), while the GSH levels in the liver were significantly lower (P<0.05). These indices improved significantly in the treatment group compared with the model group (P<0.05). In conclusion, glutamine reduces the degree of oxidative stress in the liver, inhibits NF-κB p65 expression and improves hepatic steatosis. Glutamine has a certain protective effect in NAFLD.
Collapse
Affiliation(s)
- Zhihui Lin
- Department of Gastroenterology, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Fangfang Cai
- Department of Gastroenterology, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Ning Lin
- Department of Ultrasound, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Jinli Ye
- Department of Gastroenterology, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Qiqi Zheng
- Department of Gastroenterology, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Guisheng Ding
- Department of Ultrasound, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
45
|
Salman AE, Yetişir F, Kılıç M, Onal O, Dostbil A, Zeybek D, Aksoy M, Kaymak F, Celik T, Unver S. The impact of pretreatment with bolus dose of enteral glutamine on acute lung injury induced by oleic acid in rats. J Anesth 2013; 28:354-62. [PMID: 24271996 DOI: 10.1007/s00540-013-1745-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/30/2013] [Indexed: 01/29/2023]
Abstract
PURPOSE Both parenteral and enteral glutamine have shown beneficial effects in sepsis and ischemia/reperfusion-induced acute lung injury (ALI). Oleic acid (OA) has been used to induce ALI in experimental studies. In this study, we investigated the effects of pretreatment of a bolus dose of enteral glutamine on ALI induced by OA in rats. METHODS Twenty-eight adult female Sprague-Dawley rats weighing 240-300 g were divided into four groups, 7 in each. Group I and group II received normal saline for 30 days, group III and group IV received glutamine at a dose of 1 g/kg for 10 days by gavage, and in group II and group IV 100 mg/kg OA was administered i.v. Histopathological examination of the lung was performed with light and electron microscopy. Levels of protein carbonyl, malondialdehyde, superoxide dismutase, catalase, and glutathione peroxidase levels were measured in tissue samples. Levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-10, and total tissue oxidant status and total tissue antioxidant status were measured in serum samples. RESULTS Light microscopy showed that the total lung injury score of group IV was significantly lower than group II. Change in thickness of the fused basal lamina was not significantly different in groups II and IV under electron microscopy. TNF-α, IL-6, and IL-10 serum levels were higher in group II when compared to group I and significantly attenuated in group IV. CONCLUSION Pretreatment with a bolus dose of enteral glutamine minimized the extent of ALI induced by OA in rats.
Collapse
Affiliation(s)
- A Ebru Salman
- Anesthesiology and Reanimation Department, Atatürk Research and Training Hospital, Ankara, Turkey
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Pierre JF, Heneghan AF, Lawson CM, Wischmeyer PE, Kozar RA, Kudsk KA. Pharmaconutrition Review. JPEN J Parenter Enteral Nutr 2013; 37:51S-65S. [DOI: 10.1177/0148607113493326] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Joseph F. Pierre
- Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison
| | - Aaron F. Heneghan
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison
| | - Christy M. Lawson
- Department of Surgery, University of Tennessee Medical Center, Knoxville
| | | | - Rosemary A. Kozar
- Department of Surgery, University of Texas–Houston Health Science Center, Houston
| | - Kenneth A. Kudsk
- Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison
| |
Collapse
|
47
|
Alanyl-glutamine resolves lipopolysaccharide-induced lung injury in mice by modulating the polarization of regulatory T cells and T helper 17 cells. J Nutr Biochem 2013; 24:1555-63. [DOI: 10.1016/j.jnutbio.2013.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 01/07/2013] [Accepted: 01/11/2013] [Indexed: 12/22/2022]
|
48
|
Glutamine and alanyl-glutamine increase RhoA expression and reduce Clostridium difficile toxin-a-induced intestinal epithelial cell damage. BIOMED RESEARCH INTERNATIONAL 2012; 2013:152052. [PMID: 23484083 PMCID: PMC3591182 DOI: 10.1155/2013/152052] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/12/2012] [Indexed: 12/18/2022]
Abstract
Clostridium difficile is a major cause of antibiotic-associated colitis and is associated with significant morbidity and mortality. Glutamine (Gln) is a major fuel for the intestinal cell population. Alanyl-glutamine (Ala-Gln) is a dipeptide that is highly soluble and well tolerated. IEC-6 cells were used in the in vitro experiments. Cell morphology was evaluated by atomic force microscopy (AFM) and scanning electron microscopy (SEM). Cell proliferation was assessed by WST-1 and Ki-67 and apoptosis was assessed by TUNEL. Cytoskeleton was evaluated by immunofluorescence for RhoA and F-actin. RhoA was quantified by immunoblotting. TcdA induced cell shrinkage as observed by AFM, SEM, and fluorescent microscopy. Additionally, collapse of the F-actin cytoskeleton was demonstrated by immunofluorescence. TcdA decreased cell volume and area and increased cell height by 79%, 66.2%, and 58.9%, respectively. Following TcdA treatment, Ala-Gln and Gln supplementation, significantly increased RhoA by 65.5% and 89.7%, respectively at 24 h. Ala-Gln supplementation increased cell proliferation by 137.5% at 24 h and decreased cell apoptosis by 61.4% at 24 h following TcdA treatment. In conclusion, TcdA altered intestinal cell morphology and cytoskeleton organization, decreased cell proliferation, and increased cell apoptosis. Ala-Gln and Gln supplementation reduced intestinal epithelial cell damage and increased RhoA expression.
Collapse
|
49
|
Chamney C, Godar M, Garrigan E, Huey KA. Effects of glutamine supplementation on muscle function and stress responses in a mouse model of spinal cord injury. Exp Physiol 2012; 98:796-806. [PMID: 23143993 DOI: 10.1113/expphysiol.2012.069658] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Spinal cord injury (SCI) results in loss of muscle function due to rapid breakdown of contractile proteins. Glutamine supplementation improves clinical outcomes, but its effects on muscle function after SCI are unknown. The benefits of glutamine in non-skeletal muscle tissues involve elevated heat shock protein (Hsp)70 and Hsp25, but the muscle response may differ because it is the largest contributor to plasma glutamine. We tested the hypothesis that glutamine preserves muscle function after SCI and that this is associated with increased heat shock protein and reduced inflammatory factors, interleukin-6 (IL-6) and tumour necrosis factor-α (TNFα). Changes in plantarflexor force, fatigability and total myofibrillar, Hsp70, Hsp25, IL-6 and TNFα muscle protein levels were measured 7 days after sham or spinal cord transection surgery in mice receiving daily placebo or glutamine. Compared with placebo, after SCI glutamine significantly attenuated the reductions in maximal isometric force (0.22 ± 0.01 versus 0.31 ± 0.03 N, respectively) and fatigue resistance (34 ± 4 versus 59 ± 4% of initial force, respectively). Glutamine significantly ameliorated the loss of myofibrillar protein with spinal cord transection. Spinal cord transection was associated with decreased Hsp70 and Hsp25 with glutamine only (45 ± 3 and 44 ± 5% of placebo, respectively). Glutamine significantly reduced spinal cord transection-associated increases in IL-6 and TNFα compared with placebo (38 ± 6 and 37 ± 8% of placebo, respectively). Functionally, early reductions in contractile protein, force and fatigue resistance after SCI were reversed with glutamine. Spinal cord transection-associated reductions in Hsp70, Hsp25, IL-6 and TNFα with glutamine versus placebo suggest lower stress in the muscle, possibly related to a reduced need to produce glutamine. These findings support glutamine as a therapeutic intervention to accelerate recovery of muscle function after SCI.
Collapse
Affiliation(s)
- Carissa Chamney
- College of Pharmacy and Health Sciences, Drake University, 2507 University Avenue, Des Moines, IA 50311, USA
| | | | | | | |
Collapse
|
50
|
Glutamine randomized studies in early life: the unsolved riddle of experimental and clinical studies. Clin Dev Immunol 2012; 2012:749189. [PMID: 23019424 PMCID: PMC3457673 DOI: 10.1155/2012/749189] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 07/25/2012] [Indexed: 11/21/2022]
Abstract
Glutamine may have benefits during immaturity or critical illness in early life but its effects on outcome end hardpoints are controversial. Our aim was to review randomized studies on glutamine supplementation in pups, infants, and children examining whether glutamine affects outcome. Experimental work has proposed various mechanisms of glutamine action but none of the randomized studies in early life showed any effect on mortality and only a few showed some effect on inflammatory response, organ function, and a trend for infection control. Although apparently safe in animal models (pups), premature infants, and critically ill children, glutamine supplementation does not reduce mortality or late onset sepsis, and its routine use cannot be recommended in these sensitive populations. Large prospectively stratified trials are needed to better define the crucial interrelations of “glutamine-heat shock proteins-stress response” in critical illness and to identify the specific subgroups of premature neonates and critically ill infants or children who may have a greater need for glutamine and who may eventually benefit from its supplementation. The methodological problems noted in the reviewed randomized experimental and clinical trials should be seriously considered in any future well-designed large blinded randomized controlled trial involving glutamine supplementation in critical illness.
Collapse
|