Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
ACS Appl Mater Interfaces ; 12(37): 41127-41137, 2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32808767

ABSTRACT

Weak T cell responses and immune checkpoints within tumors could be two key factors for limiting antitumor efficacy in the field of cancer immunotherapy. Thus, the combined strategy of tumor vaccines and immune checkpoint blockade has been widely studied and expected to boost antitumor immune responses. Herein, we first developed a two-barreled strategy to combine the nanovaccine with a gene-mediated PD-L1 blockade. On the one hand, polyethyleneimine (PEI) worked as a vaccine carrier to codeliver the antigen ovalbumin (OVA) and the adjuvant unmethylated cytosine-phosphate-guanine (CpG) to formulate the PEI/OVA/CpG nanovaccine through electrostatic binding, which realized both dendritic cell activation and antigen cross-presentation enhancement. On the other hand, the PD-L1 silence gene was loaded by PEI to form PEI/pshPD-L1 complexes, which were further in situ shielded by aldehyde-modified polyethylene glycol (OHC-PEG-CHO) via pH-responsive Schiff base bonds. The formed pshPD-L1@NPs could decrease PD-L1 expression on the tumor cells. However, such a combined two-barreled strategy improved feebly for tumor inhibition in comparison with monotherapy, exhibiting the antagonistic effect, which might be due to the limited T cell response enhancement in the tumor microenvironment. To solve this problem, we have further developed a three-barreled strategy to combine oral administration of l-arginine, which worked as an amplifier to induce robust T cell response enhancement, without causing the upregulation of other negative immune regulators. Superior antitumor behavior and tumor rechallenge protection were realized by the three-barreled strategy in B16F10-OVA (B16-OVA)-bearing mice. The unique three-barreled strategy we developed might offer a novel clinical therapeutic treatment.


Subject(s)
Arginine/immunology , B7-H1 Antigen/antagonists & inhibitors , Cancer Vaccines/immunology , Immunotherapy , Nanoparticles/chemistry , Animals , Arginine/chemistry , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cancer Vaccines/chemistry , Cytosine/chemistry , Cytosine/immunology , Guanine/chemistry , Guanine/immunology , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Ovalbumin/chemistry , Ovalbumin/immunology , Particle Size , Phosphates/chemistry , Phosphates/immunology , Polyethyleneimine/chemistry , Surface Properties
2.
Biomed Res ; 40(1): 1-7, 2019.
Article in English | MEDLINE | ID: mdl-30787259

ABSTRACT

For a long time, many types of vaccines have been useful for the prophylaxis of many infectious diseases. Thus far, many adjuvants that enhance the effects of vaccines have been explored. However, very few adjuvants are being used for humans worldwide. In this study, we investigated the adjuvant activity of various substances, and found citrulline to have high potential as an adjuvant. Citrulline is a type of amino acid present in the body of many organisms. A number of biological activities of citrulline have been reported; however, no adjuvant activity has been reported thus far. Aluminum salts, which are commonly used as adjuvants are not water soluble; therefore, some difficulties are encountered while using them as vaccine adjuvants. Citrulline is easy to use because of its water solubility. In this study, we showed for the first time the adjuvant activity of citrulline by using viral antigens and amyloid ß peptide. Water-soluble citrulline, which is present in our body, is a potential adjuvant candidate.


Subject(s)
Adjuvants, Immunologic , Citrulline , Vaccines , Alum Compounds , Amyloid beta-Peptides/immunology , Animals , Arginine/immunology , Citrulline/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunization , Mice , Peptides/immunology , Vaccines/immunology , Vaccines, Inactivated
3.
J Surg Oncol ; 115(3): 273-280, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27861915

ABSTRACT

Arginine is an important player in numerous biologic processes and studies have demonstrated its importance for cellular growth that becomes limiting in states of rapid turnover (e.g., malignancy). Thus, arginine deprivation therapy is being examined as an adjuvant cancer therapy, however, arginine is also necessary for immune destruction of malignant cells. Herein we review the data supporting arginine deprivation or supplementation in cancer treatment and the currently registered trials aimed at understanding these divergent strategies. J. Surg. Oncol. 2017;115:273-280. © 2016 Wiley Periodicals, Inc.


Subject(s)
Arginine/administration & dosage , Arginine/deficiency , Neoplasms/therapy , Animals , Arginine/immunology , Arginine/metabolism , Dietary Supplements , Humans , Neoplasms/immunology , Neoplasms/metabolism
4.
Pediatr Res ; 77(2): 290-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25360828

ABSTRACT

Preterm newborns are highly susceptible to bacterial infections. This susceptibility is regarded as being due to immaturity of multiple pathways of the immune system. However, it is unclear whether a mechanism that unifies these different, suppressed pathways exists. Here, we argue that the immune vulnerability of the preterm neonate is critically related to arginine depletion. Arginine, a "conditionally essential" amino acid, is depleted in acute catabolic states, including sepsis. Its metabolism is highly compartmentalized and regulated, including by arginase-mediated hydrolysis. Recent data suggest that arginase II-mediated arginine depletion is essential for the innate immune suppression that occurs in newborn models of bacterial challenge, impairing pathways critical for the immune response. Evidence that arginine depletion mediates protection from immune activation during first gut colonization suggests a regulatory role in controlling gut-derived pathogens. Clinical studies show that plasma arginine is depleted during sepsis. In keeping with animal studies, small clinical trials of L-arginine supplementation have shown benefit in reducing necrotizing enterocolitis in premature neonates. We propose a novel, broader hypothesis that arginine depletion during bacterial challenge is a key factor limiting the neonate's ability to mount an adequate immune response, contributing to the increased susceptibility to infections, particularly with respect to gut-derived sepsis.


Subject(s)
Arginine/deficiency , Bacterial Infections/etiology , Disease Susceptibility/immunology , Immunity, Innate/immunology , Infant, Premature/immunology , Microbiota/immunology , Models, Immunological , Arginine/immunology , Arginine/metabolism , Bacterial Infections/immunology , Gastrointestinal Tract/microbiology , Humans , Infant, Newborn , Signal Transduction/immunology
5.
Asia Pac J Clin Nutr ; 23(3): 351-9, 2014.
Article in English | MEDLINE | ID: mdl-25164444

ABSTRACT

L-arginine plays an important role in immune regulation by affecting the immune response and inflammation. This meta-analysis was performed to assess whether L-arginine supplementation could improve the outcomes of immune function, and to evaluate the safety of L-arginine supplementation. Four databases (PubMed, EMBASE, Web of Science, the Cochrane Library) for all randomized controlled trials investigating the effects of supplementation with L-arginine published from 1966 to September 2013 were searched. The quality of controlled trials was assessed with the Jadad method. Meta-analyses were performed with fixed- or random-effects models according to heterogeneity of studies. Data from 11 trials involving 321 patients were enrolled. Meta-analysis showed that the L-arginine supplement group had a significantly greater CD4⁺ T-cell proliferation response (MD 5.03; 95% CI 1.11, 8.95; p<0.05), and that the incidence of infectious complications was lower (OR 0.40; 95% CI 0.17, 0.95; p<0.05) than control.


Subject(s)
Arginine/immunology , Arginine/pharmacology , Dietary Supplements , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Cytokines/immunology , Humans , Randomized Controlled Trials as Topic/methods
6.
Int Immunopharmacol ; 16(2): 322-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23619555

ABSTRACT

Endothelial dysfunction is the early stage of atherosclerosis, which is typically associated with rheumatoid arthritis (RA), a chronic inflammatory autoimmune disorder. Asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase inhibitor, is not only an independent predictor for endothelial dysfunction but also a proinflammatory mediator. It has been shown that the level of ADMA was elevated in patients with RA. In the present study, we investigated the potential effect of ADMA on inflammation process in collagen-induced arthritis (CIA) animal model and primary cultured fibroblast-like synoviocytes (FLS) exposed to tumor necrosis factor-α (TNF-α). In CIA rats, the plasma levels of inflammatory cytokines TNF-α, interleukin-1ß (IL-1ß) and IL-6 were markedly increased, while the plasma levels of ADMA did not increase. The expression of dimethylarginine dimethylohydrolase2 (DDAH2), the key enzyme for ADMA degradation, was markedly reduced in inflamed joint synovium of CIA rats. Moreover, the expression of anti-inflammatory factor cortistatin (CST) was markedly decreased in joint synovium of CIA rats. Treatment of cultured FLS with TNF-α significantly increased the levels of ADMA, and decreased the expression of DDAH2 mRNA and protein accompany with an increase in the levels of IL-1ß and IL-6 and a reduction in the expression of CST mRNA and protein, and the effects of TNF-α were abolished by DDAH2 overexpression. Treatment of FLS with ADMA also significantly increased the levels of IL-1ß and IL-6, and reduced the expression of CST. These findings suggest that DDAH/ADMA participates in the pathogenesis of RA, and that the effect of DDAH/ADMA may be mediated by CST.


Subject(s)
Amidohydrolases/immunology , Arginine/analogs & derivatives , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Neuropeptides/immunology , Amidohydrolases/genetics , Animals , Ankle Joint/pathology , Arginine/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Cytokines/blood , Cytokines/genetics , Male , Rats , Rats, Sprague-Dawley , Synovial Membrane/immunology , Synovial Membrane/pathology
7.
BMC Complement Altern Med ; 13: 274, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-24499201

ABSTRACT

BACKGROUND: Injury to a nerve is the most common reason of acquired peripheral neuropathy. Therefore, searching for effective substance to recover of nerve after injury is need of present era. The current study investigates the protective potential of Standardized Fruit Extract of Punica granatum L (PFE) [Ellagic acid (41.6%), Punicalagins (10%), Granatin (5.1%)] in Tibial & Sural Nerve Transection (TST) induced neuropathic pain in rats. METHODS: TST was performed by sectioning tibial and sural nerve portions of the sciatic nerve and leaving the common peroneal nerve intact. Acetone drop, pin-prick, hot plate, paint brush & Walking Track tests were performed to assess cold allodynia; mechanical heat, hyperalgesia and dynamic mechanical allodynia & tibial functional index respectively. The levels of TNF-α, TBARS, GSH and Nitrite were measured in the sciatic nerve as an index of inflammation & oxidative stress. RESULTS: TST led to significant development of cold allodynia; mechanical and heat hyperalgesia; dynamic mechanical allodynia; functional deficit in walking along with rise in the levels of TBARS, TNF-α, GSH and Nitrite. Administrations of PFE (100 & 300 mg/kg oral), significantly attenuate TST induced behavioral & biochemical changes. Pretreatments of BADGE (120 mg/kg IP) a PPAR-γ antagonist and nitric oxide precursor L-arginine (100 mg/kg IP) abolished the protective effect of PFE. Whereas, pretreatment of L-NAME (5 mg/kg IP) a NOS inhibitor significantly potentiated PFE's protective effect of PFE. CONCLUSION: PFE shown to have attenuating effect in TST induced neuropathic pain which may be attributed to potential PPAR-gamma agonistic activity, nitric oxide inhibitory, anti-inflammatory and anti oxidative actions.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Lythraceae/chemistry , Neuralgia/drug therapy , Plant Extracts/administration & dosage , Sural Nerve/injuries , Tibial Nerve/injuries , Animals , Arginine/immunology , Female , Fruit/chemistry , Humans , Male , Neuralgia/genetics , Neuralgia/immunology , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sural Nerve/drug effects , Tibial Nerve/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
8.
Am J Surg ; 204(4): 416-21, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23010614

ABSTRACT

AIM: The importance of the alteration of tumor infiltrative lymphocytes (CD4(+), CD8(+), CD16(+), and CD56(+)) in colorectal cancer prognosis is well known. In this study, we analyzed the effect of preoperative immunonutrition and different nutritional models on the clinical condition of colorectal cancer patients. METHODS: Twenty-eight colorectal cancer patients were grouped into 4 groups according to their nutrition regimens randomly and were given immunonutrition (IMN), standard enteral (SE), total parental nutrition (TPN), and normal nutrition (NN) regimens, all of which contained the same calorie-nitrogen content within a 7-day preoperative period. All patients had an endoscopic biopsy before and after the regimen, and the lymphocyte population infiltrating mucosal parts of the resected tumor tissue were evaluated. Immunohistochemical analysis of the tissue specimens was performed by staining with antihuman CD4(+), CD8(+), CD16(+), and CD56(+) antibodies. RESULTS: After nutrition, there were significant increases in each of the 4 groups of CD4(+) and CD8(+) cells within the tumor. Comparing the rates of augmentation, the increased rates of the CD8(+) cells infiltrating the tumor after nutrition in the patients who were fed with IMN were significantly more than the ones in other groups (P = .01). CD16(+) cell infiltration was significantly higher in all groups except the SE and IMN groups. The SE group had increased CD56(+) cell infiltration compared with the other groups. CONCLUSIONS: In the colorectal cancer patients who had nutrition in the 7-day preoperative period, except for the SE nutrition group, there were significant increases of infiltration of CD56(+) cells at the mucosal part of the tumor tissue within the CD4(+) and CD8(+) cell population. When postnutrition values were compared, there was a marked increase of CD8(+) cells in the IMN group.


Subject(s)
Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Enteral Nutrition , Food, Formulated , Lymphocytes, Tumor-Infiltrating/immunology , Parenteral Nutrition, Total , Aged , Arginine/administration & dosage , Arginine/immunology , Biopsy , CD4-Positive T-Lymphocytes/immunology , CD56 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Colonoscopy , Dietary Proteins/administration & dosage , Dietary Proteins/immunology , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/immunology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Nutritional Physiological Phenomena , Preoperative Period , RNA/administration & dosage , RNA/immunology , Receptors, IgG/immunology , Turkey
9.
JPEN J Parenter Enteral Nutr ; 36(6): 741-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22269898

ABSTRACT

BACKGROUND: Arginine deficiency and chronic inflammation may cause immune dysfunction. The authors previously showed that a pharmacological dose of parenteral arginine facilitates ornithine rather than nitric oxide production in subacute peritonitis. Herein, they investigated the effects of different doses of parenteral arginine supplementation on immunocytic subpopulation distribution and function. MATERIALS: Male Wistar rats that underwent cecal punctures for induction of subacute peritonitis were infused with conventional parenteral nutrition solution (1.61% of total calories as arginine) or solutions supplemented with low-, medium-, or high-dose arginine (2.85%, 4.08%, and 6.54% of total calories, respectively) for 7 days. Distributions of T cells, B cells, and monocytes/macrophages and cytokine productions of peripheral blood lymphocytes (PBLs) and splenocytes were analyzed. RESULTS: There were no significant differences in circulating white blood cell numbers and serum tumor necrosis factor (TNF)-α and interferon (IFN)-γ concentrations among groups. Serum nitrate/nitrite (NOx) and interleukin (IL)-2 levels were significantly decreased by arginine in a dose-dependent manner. Animals supplemented with parenteral arginine had significantly decreased productions of concanavalin (Con) A- and lipopolysaccharide (LPS)-stimulated TNF-α in PBLs and splenocytes, spontaneous IL-6 and LPS-stimulated IFN-γ in PBLs, and LPS-stimulated IL-6 in splenocytes. In addition, low-dose arginine significantly increased production of spontaneous IFN-γ in PBLs and splenocytes. High-dose arginine significantly increased spontaneous TNF-α, and Con A stimulated IL-4 and IL-6 in PBLs. CONCLUSION: Parenteral arginine administration at approximately 4% of total calories may alter PBLs and splenocytic immunity, and >6% of total calories might not be of benefit in rats with subacute peritonitis.


Subject(s)
Arginine/administration & dosage , Deficiency Diseases , Immunity/drug effects , Inflammation Mediators/metabolism , Inflammation/drug therapy , Leukocytes/drug effects , Peritonitis/drug therapy , Animals , Arginine/deficiency , Arginine/immunology , Arginine/therapeutic use , Chronic Disease , Concanavalin A/blood , Cytokines/metabolism , Deficiency Diseases/complications , Deficiency Diseases/drug therapy , Deficiency Diseases/immunology , Dietary Supplements , Dose-Response Relationship, Drug , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharides , Macrophages/drug effects , Male , Nitrates/blood , Nitrites/blood , Parenteral Nutrition , Peritonitis/complications , Peritonitis/immunology , Peritonitis/therapy , Rats , Rats, Wistar , Spleen/cytology , Spleen/drug effects , Spleen/immunology
10.
ScientificWorldJournal ; 11: 2443-57, 2011.
Article in English | MEDLINE | ID: mdl-22219714

ABSTRACT

Dietary supplementation with L-arginine was shown to improve immune responses in various inflammatory models. However, the molecular mechanisms underlying L-arginine effects on immune cells remain unrecognized. Herein, we tested the hypothesis that a limitation of L-arginine could lead to the uncoupled state of murine macrophage inducible nitric oxide synthase and, therefore, increase inducible nitric-oxide-synthase-derived superoxide anion formation. Importantly, we demonstrated that L-arginine dose- and time dependently potentiated superoxide anion production in bacterial endotoxin-stimulated macrophages, although it did not influence NADPH oxidase expression and activity. Detailed analysis of macrophage activation showed the time dependence between LPS-induced iNOS expression and increased O(2)(∙-) formation. Moreover, downregulation of macrophage iNOS expression, as well as the inhibition of iNOS activity by NOS inhibitors, unveiled an important role of this enzyme in controlling O(2)(∙-) and peroxynitrite formation during macrophage stimulation. In conclusion, our data demonstrated that simultaneous induction of NADPH oxidase, together with the iNOS enzyme, can result in the uncoupled state of iNOS resulting in the production of functionally important levels of O(2)(∙-) soon after macrophage activation with LPS. Moreover, we demonstrated, for the first time that increased concentrations of L-arginine further potentiate iNOS-dependent O(2) (∙-) formation in inflammatory macrophages.


Subject(s)
Arginine/immunology , Macrophages/immunology , Nitric Oxide Synthase Type II/metabolism , Superoxides/metabolism , Animals , Biopterins/analogs & derivatives , Biopterins/metabolism , Cell Line , Cell Survival , Enzyme Activation , Enzyme Inhibitors/pharmacology , Escherichia coli/immunology , Lipopolysaccharides/adverse effects , Macrophages/drug effects , Macrophages/metabolism , Mice , NADPH Oxidases/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Respiratory Burst , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism
11.
Rio de Janeiro; s.n; 2011. 111 p. ilus, tab.
Thesis in Portuguese | LILACS | ID: lil-613885

ABSTRACT

A L-aginina é reconhecida como um nutriente de fundamental importância na resposta imune, apesar de seus efeitos serem, por vezes, considerados inconstantes. O autoimplante esplênico tem sido proposto como alternativa à esplenectomia total isolada, mas existem preocupações quanto à eficácia do restabelecimento da resposta imune, haja vista que o paciente pode permanecer com risco aumentado de desenvolvimento de infecção fulminante pós esplenectomia, mesmo após a regeneração morfológica do órgão. O objetivo deste estudo foi avaliar a participação da suplementação dietética com L-arginina em subpopulações linfocitárias no sangue, no baço e nos autoimplantes esplênicos de ratos submetidos a esplenectomia isolada ou combinada com autoimplante esplênico. Foram utilizados 42 ratos Sprague-Dawlay machos, randomicamente distribuídos em seis grupos: 1 - Controle - operação simulada; 2 - esplenectomia total; 3 - esplenectomia total combinada com autoimplante esplênico; 4 - Controle - operação simulada, com suplementação de L-arginina; 5 - esplenectomia total, com suplementação de L-arginina; e 6 - esplenectomia total combinada com autoimplante esplênico, com suplementação de L-arginina. Os animais dos grupos 4, 5 e 6 receberam suplementação de L-arginina, uma vez ao dia, durante 15 dias anteriores a coleta sangüínea realizada imediatamente antes dos procedimentos operatórios (semanas 0 e 12). A dose utilizada foi de 1,0 g/kg/dia, administrada por via intragástrica em bolus. As avaliações foram realizadas por meio de hemograma e citometria de fluxo. A análise estatística utilizou testes paramétricos e não-paramétricos, sendo p<0,05 considerado para a rejeição da hipótese nula. A suplementação com L-arginina acarretou elevação da contagem relativa e absoluta de neutrófilos periféricos, 12 semanas após a realização de esplenectomia total combinada com autoimplante esplênico. A esplenectomia total ocasionou diminuição da contagem relativa de linfócitos T totais, T CD4+...


L-arginine is recognized as a nutrient of fundamental importance in immune implants has been proposed as an alternative to total splenectomy isolated, but there are concerns about the effectiveness of the restoration of the immune response, considering that the patient can remain at increased risk of developing overwhelming postsplenectomy infection, even after morphological regeneration of the organ. The aim of this study was to determine the role of dietary supplementation with L-arginine in lymphocyte subsets in blood, spleen, and splenic auto-transplantation in rats subjected to total splenectomy alone or in combination with splenic auto-transplantation. Forty two male Sprague-Dawley rats, were randomly divided into six groups: 1 - Control - sham operation, 2 - total splenectomy, 3 - total splenectomy combined with splenic auto-implants, 4 - Control - sham operation, with L-arginine supplementation, 5 - total splenectomy, supplemented with L-arginine, and 6 - total splenectomy combined with splenic auto-implants, supplemented with L-arginine. Animals in groups 4, 5 and 6 were supplemented with L-arginine, once daily for 15 days before blood sample was collected immediately before the operative procedures (weeks 0 and 12). The dose was 1.0 g/kg/day administered by intragastric bolus. The laboratory evaluations were made by blood count and flow cytometry. Statistical analysis used parametric tests and nonparametric, p<0.05 was considered to reject the null hypothesis. Supplementation with L-arginine led to increase in relative and absolute count of peripheral neutrophils, 12 weeks after completion of total splenectomy combined with splenic auto-implants. Total splenectomy caused a decrease in relative count of T lymphocytes, CD4+ and CD8B in blood, but dietary supplementation with L-arginine prevented the decrease in the percentage of total T cells and CD8B in the blood of animals subjected to splenic auto-transplantation...


Subject(s)
Rats , Arginine/administration & dosage , Arginine/immunology , Spleen/immunology , Spleen/transplantation , Splenectomy/methods , Immune System , Lymphocyte Subsets , Lymphocyte Subsets/immunology , Dietary Supplements , Transplantation, Autologous , Infections/etiology , Lymphocytes/immunology
12.
Clin Nutr ; 29(5): 654-62, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20392549

ABSTRACT

BACKGROUND & AIMS: Immune-enhancing diets (IEDs) contain a mixture of nutrients claimed to have immunological properties. Therefore, it seemed relevant to determine the effect of each of their components. The aim of this study was to examine the role of arginine (Arg) and ω3 polyunsaturated fatty acids (ω3 PUFAs) in the effect of an IED (Crucial(®)) in a validated rat model of inflammation induced by turpentine (TI). METHODS: Forty-two rats were randomized into five groups: AL (ad libitum), TI-EN (TI+standard enteral nutrition (EN): Sondalis(®)HP), TI-EN-Arg (TI+standard EN+Arg in equimolar concentration to Arg in the IED), TI-M-IED (TI+modified IED containing the same ω6/ω3 ratio as in standard EN) and TI-IED (TI+Crucial(®)). Blood was sampled to determine CD25 receptor density on lymphocytes. TNF-α, IL-6 and NO (production and expression) were evaluated on isolated macrophages. Mesenteric lymph nodes, spleen and liver were cultured for analysis of enterobacterial translocation and dissemination. RESULTS: CD25 density was decreased after TI and was corrected in the TI-EN-Arg, TI-M-IED and TI-IED groups (p<0.05). TI induced an alteration of macrophage mRNA expression of IL-6, TNF-α and iNOS, corrected in the TI-EN-Arg and TI-M-IED groups (p<0.05), but not by the IED. Enterobacterial translocation was observed in all treated groups, nevertheless the amount tended (p=0.054) to be lower in the TI-EN-Arg group. CONCLUSIONS: Arg and ω3 PUFAs make a major contribution to IED effects, but our study shows interaction between them on macrophage reactivity. This indicates that the individual properties of each pharmaconutrient are not additive in IEDs.


Subject(s)
Arginine/pharmacokinetics , Fatty Acids, Omega-3/pharmacokinetics , Food, Formulated , Inflammation/metabolism , Inflammation/therapy , Animals , Arginine/immunology , Bacterial Translocation , Drug Interactions , Enteral Nutrition , Fatty Acids, Omega-3/immunology , Interleukin-2 Receptor alpha Subunit/analysis , Interleukin-2 Receptor alpha Subunit/blood , Interleukin-6/immunology , Interleukin-6/metabolism , Lymphocytes/metabolism , Male , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Turpentine/metabolism
13.
World J Surg ; 33(9): 1815-21, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19629583

ABSTRACT

BACKGROUND: Past trials have shown perioperative immunonutrition to improve the outcome for patients with gastric cancer. The present study was designed to evaluate the effect of preoperative oral immunonutrition on cellular immunity, the duration of the systemic inflammatory response syndrome (SIRS), and detailed postoperative complications in patients with gastric cancer. METHODS: Sixty patients with gastric cancer were randomly assigned to two groups: one group received immune-enhanced formulas supplemented with arginine and omega-3 fatty acids (immune-enhancing diet (ID) group, n = 30); the other received standard formulas (conventional diet (CD) group, n = 30) for 7 days before the operation. These groups were well matched in terms of age, sex, operations, cancer stages, and intraoperative variables. The postoperative outcome was evaluated based on clinical variables, including postoperative infectious complications, noninfectious complications, and SIRS duration. In addition, the perioperative state of cellular immunity was evaluated and compared between the two groups. RESULTS: The incidence of postoperative infectious complications in the ID group (6%) was significantly (p < 0.05) lower than that of the CD group (28%). The duration of SIRS in the ID group (0.77 +/- 0.9 days) was significantly (p < 0.05) shorter than that in the CD group (1.34 +/- 1.45 days). The postoperative lymphocyte and CD4(+)T-cell counts significantly decreased (p < 0.05) in both groups. However, the number of CD4(+)T-cells on preoperative day 1 and postoperative day 7 was significantly (p < 0.05) higher in the ID group than in the CD group. CONCLUSIONS: Preoperative oral immune-enhanced formulas supplemented with arginine and omega-3 fatty acids enhanced the immune status of the patients, reduced the duration of SIRS, and decreased the incidence of postoperative infectious complications. CD4(+)T-cell immunity likely played an important role in the modulation of the postoperative immune and inflammatory response after gastrectomy.


Subject(s)
Arginine/immunology , Fatty Acids, Omega-3/immunology , Gastrectomy , Immunity, Cellular/drug effects , Infections/immunology , Postoperative Complications/immunology , Postoperative Complications/prevention & control , Preoperative Care , Stomach Neoplasms/surgery , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/prevention & control , Administration, Oral , Adult , Aged , Aged, 80 and over , Analysis of Variance , Arginine/administration & dosage , Chi-Square Distribution , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Female , Humans , Incidence , Infections/epidemiology , Lymphocyte Count , Male , Middle Aged , Postoperative Complications/epidemiology , Statistics, Nonparametric , Stomach Neoplasms/immunology , Systemic Inflammatory Response Syndrome/epidemiology , Treatment Outcome
14.
Rev. Nac. (Itauguá) ; 1(2): 31-38, 2009.
Article in Spanish | LILACS, BDNPAR | ID: biblio-1017760

ABSTRACT

La suplementación de la nutrición enteral con Arginina a dosis mínima (6 g. día) desde el primer día de alimentación posterior a una cirugía abdominal complicada tendría efectos sobre la respuesta inmunológica y los niveles de proteínas plasmáticas. Se encontró un aumento significativo del Recuento Total de Linfocitos y de los niveles de Transferrina en el grupo que recibió la Arginina. Las demás variables no tuvieron una variación significativa...


Subject(s)
Humans , Food, Formulated , Nutritional Support , Arginine/administration & dosage , Arginine/immunology , Arginine/therapeutic use , Enteral Nutrition , Immune System
15.
J Nutr ; 137(6 Suppl 2): 1681S-1686S, 2007 06.
Article in English | MEDLINE | ID: mdl-17513447

ABSTRACT

For many years, dietary arginine supplementation, often combined with other substances, has been used as a mechanism to boost the immune system. Considerable controversy, however, exists as to the benefits and indications of dietary arginine due in part to a poor understanding of the role played by this amino acid in maintaining immune function. Emerging knowledge promises to clear this controversy and allow for arginine's safe use. In myeloid cells, arginine is mainly metabolized either by inducible nitric oxide (NO) synthases (iNOS) or by arginase 1, enzymes that are stimulated by T helper 1 or 2 cytokines, respectively. Thus, activation of iNOS or arginase (or both) reflects the type of inflammatory response in a specific disease process. Myeloid suppressor cells (MSC) expressing arginase have been described in trauma (in both mice and humans), intra-abdominal sepsis, certain infections, and prominently, cancer. Myeloid cells expressing arginase have been shown to accumulate in patients with cancer. Arginase 1 expression is also detected in mononuclear cells after trauma or surgery. MSC efficiently deplete arginine and generate ornithine. Through arginine depletion, MSC may control NO production and regulate other arginine-dependent biological processes. Low circulating arginine has been documented in trauma and cancer, suggesting that MSC may exert a systemic effect and cause a state of arginine deficiency. Simultaneously, T lymphocytes depend on arginine for proliferation, zeta-chain peptide and T-cell receptor complex expression, and the development of memory. T-cells cocultured with MSC exhibit the molecular and functional effects associated with arginine deficiency. Not surprisingly, T-cell abnormalities, including decreased proliferation and loss of the zeta-chain, are observed in cancer and after trauma.


Subject(s)
Arginine/immunology , Arginine/metabolism , Immune System/metabolism , Myeloid Cells/metabolism , T-Lymphocytes/metabolism , Animals , Humans , Immune System/cytology , Immune System/immunology , Myeloid Cells/immunology , T-Lymphocytes/immunology
16.
J Nutr ; 137(6 Suppl 2): 1687S-1692S, 2007 06.
Article in English | MEDLINE | ID: mdl-17513448

ABSTRACT

Arginine is a nonessential amino acid in the normal physiological state that becomes conditionally essential during periods of hypermetabolic stress. Recent literature supports the hypothesis that arginine plays an important role in the intermediary metabolism of the critically ill patient. Current critical care literature is conflicting on arginine use in the clinical setting, with some proposing it as a panacea, whereas others report it as poison. Multiple individual reports and at least 5 major meta-analyses using combinations of immune-modulating nutrients have reported mostly beneficial results, but few have evaluated the effects of arginine when given as a single supplemental nutrient. This review attempts to objectively analyze the literature and evaluate the potential role of arginine in the critical care setting.


Subject(s)
Arginine/metabolism , Arginine/therapeutic use , Critical Illness , Stress, Physiological/drug therapy , Stress, Physiological/metabolism , Arginine/immunology , Critical Care/methods , Humans , Stress, Physiological/immunology
17.
J Bioinform Comput Biol ; 4(2): 415-24, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16819792

ABSTRACT

The third complementary determining region of the immunoglobulin heavy chain (CDR H3) is one of the more difficult structures to model due to genetic reasons. However, the conformation of proximal to beta-framework ("torso") part of the CDR H3 is very predictable. Current "CDR's canonical classes" theory is based on identifying the key positions, H94 and H101. We can determine the CDR H3 "torso" structure if arginine or lysine is present in the H94 position and/or aspartic acid in the H101 position. We target the case characterized by the absence of key residues in both the H94 and H101 positions. There has not been discussion on this case in the literature. 51 CDR H3 structures of this nature are analyzed and we established new sequence-structure rules. These rules contribute to more accurate modeling of the antibody's structure.


Subject(s)
Antibodies/chemistry , Immunoglobulin Heavy Chains/chemistry , Models, Chemical , Models, Molecular , Sequence Analysis, Protein/methods , Amino Acid Sequence , Amino Acid Substitution , Antibodies/immunology , Arginine/chemistry , Arginine/immunology , Aspartic Acid/chemistry , Aspartic Acid/immunology , Binding Sites , Computer Simulation , Drug Delivery Systems/methods , Immunoglobulin Heavy Chains/immunology , Models, Immunological , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Sequence Alignment/methods
19.
J Trauma ; 58(3): 455-61; discussion 461, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15761336

ABSTRACT

BACKGROUND: Recent reports suggest that enteral diets enriched with arginine may be harmful by enhancing inflammation. This is consistent with our gut ischemia/reperfusion (I/R) model in which arginine induced the proinflammatory mediator inducible nitric oxide synthase and resulted in injury and inflammation whereas glutamine was protective. We now hypothesize that arginine and glutamine differentially modulate the early proinflammatory transcription factors activated by gut I/R. METHODS: At laparotomy, jejunal sacs were filled with either 60 mmol/L glutamine, arginine, or an iso-osmotic control followed by 60 minutes of superior mesenteric artery occlusion and 6 hours of reperfusion and compared with shams. Jejunum was harvested for nuclear factor (NF)-kappaB and activator protein-1 (AP-1) measured by electrophoretic mobility shift assay and c-jun and c-fos (AP-1 family) by supershift. RESULTS: Both NF-kappaB and AP-1 were activated by gut I/R. Arginine and glutamine had no differential effect on NF-kappaB, whereas AP-1 expression (c-jun but not c-fos) was markedly enhanced by arginine and significantly lessened by glutamine. CONCLUSION: Arginine enhanced expression of the early proinflammatory transcription factor AP-1 but not NF-kappaB. This represents a novel mechanism by which arginine may be harmful when administered to critically ill patients.


Subject(s)
Arginine/therapeutic use , Enteral Nutrition/methods , Glutamine/therapeutic use , Immunologic Factors/therapeutic use , Jejunum/drug effects , Mesenteric Vascular Occlusion/therapy , NF-kappa B/drug effects , Reperfusion Injury/therapy , Transcription Factor AP-1/drug effects , Analysis of Variance , Animals , Arginine/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Electrophoretic Mobility Shift Assay , Enteral Nutrition/adverse effects , Glutamine/immunology , Immunity, Mucosal/drug effects , Immunity, Mucosal/immunology , Immunologic Factors/immunology , Inflammation , Jejunum/blood supply , Jejunum/chemistry , Jejunum/immunology , Male , Mesenteric Artery, Superior , Mesenteric Vascular Occlusion/complications , Mesenteric Vascular Occlusion/immunology , NF-kappa B/analysis , NF-kappa B/immunology , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase Type II , Patient Selection , Peroxisome Proliferator-Activated Receptors/analysis , Peroxisome Proliferator-Activated Receptors/drug effects , Peroxisome Proliferator-Activated Receptors/immunology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/complications , Reperfusion Injury/immunology , Risk Factors , Time Factors , Transcription Factor AP-1/analysis , Transcription Factor AP-1/immunology
20.
J Autoimmun ; 23(2): 141-50, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15324932

ABSTRACT

Autoreactivity is controlled at various steps by numerous mechanisms and is a key to understanding and treating autoimmune disease. Recently, an antibody against deiminated fibrinogen (DI-FBG) was detected in patients with rheumatoid arthritis (RA) with high specificity and sensitivity. DI-FBG converted enzymically by peptidyl arginine deiminase, was also detected in synovial membrane. In the present study, we investigated whether antibody to DI-FBG is produced in mice immunized with DI-FBG. Mice were immunized with DI-FBG in the presence or absence of adjuvant. Production of the specific antibody was only induced with adjuvant. The resulting antibody was specific for DI-FBG and did not react with intact/native fibrinogen. Furthermore, it recognized deiminated human fibrinogen and cyclic citrullinated peptide (CCP). These results suggested that mouse fibrinogen acquires antigenicity in mice through deimination and therefore, autoantibody such as that detected in RA patients specifically may be induced.


Subject(s)
Arginine/immunology , Autoantigens/immunology , Fibrinogen/immunology , Adjuvants, Immunologic , Animals , Antibody Formation , Arginine/metabolism , Arthritis, Rheumatoid/immunology , Autoantibodies/biosynthesis , Autoimmunity , Fibrinogen/metabolism , Hydrolases/metabolism , Imines/immunology , Mice , Mice, Inbred BALB C , Protein-Arginine Deiminases
SELECTION OF CITATIONS
SEARCH DETAIL