Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Transpl Immunol ; 27(1): 39-47, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22647882

ABSTRACT

INTRODUCTION: In this study we aimed to determine whether Castanospermine, a transplant immunosuppressive agent, impaired mononuclear/endothelial cell binding and expression of their cell adhesion molecules. METHODS: The binding of human umbilical vein endothelial cells with peripheral blood mononuclear cells was measured by a binding assay using Chromium 51 label; the membrane expression of cell adhesion molecules was measured by flow cytometry expressed as mean fluorescence intensity ratios. RESULTS: Castanospermine decreased mononuclear/endothelial cell binding if and only if both cell types were treated with Castanospermine: this impairment occurred if endothelial cells were treated with a range of doses of Castanospermine and mononuclear cells were treated with a constant dose of Castanospermine (p<0.001 versus untreated p=0.978) or vice versa (p=0.004 versus untreated p=0.582). Upon human umbilical vein endothelial cells Castanospermine reduced the mean fluorescence intensity ratios of E-selectin (p=0.003), ICAM-1 (p<0.001), ICAM-2 (p=0.004) and PECAM-1 (p<0.001) but increased it for P-selectin (p<0.001). Upon peripheral blood mononuclear cells Castanospermine reduced the mean fluorescence intensity ratios of L-selectin (P<0.001), LFA-1α (p<0.001), VLA-4 (p<0.001), Mac-1 (P<0.001) and CR4 (p<0.001) but increased the mean fluorescence intensity ratios of PSGL-1 (p<0.001) and PECAM-1 (p=0.001). Similar changes in mean fluorescence intensity ratios were found in the subset of lymphocytes and monocytes but the reductions in LFA-1α and VLA-4 on lymphocytes and Mac-1 and CR4 on monocytes were greater. CONCLUSIONS: The reduction in mononuclear/endothelial cell binding mediated by CAST and the reduction in expression of multiple cell adhesion molecules on these cell types help to explain the mechanism of its established immunosuppressive effect.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/metabolism , Cell Adhesion/drug effects , Indolizines/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , E-Selectin/metabolism , Enzyme Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunosuppressive Agents/pharmacology , Integrin alpha4beta1/metabolism , Intercellular Adhesion Molecule-1/metabolism , L-Selectin/metabolism , P-Selectin/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
2.
Travel Med Infect Dis ; 6(1-2): 67-81, 2008.
Article in English | MEDLINE | ID: mdl-18342278

ABSTRACT

It is now broadly accepted for infectious disease in general that it is not the invading organism, but the body's unbridled response to it--the "cytokine storm"--that causes illness and pathology. Nevertheless, many researchers still regard the harmful effects of falciparum malaria as being governed by oligaemic hypoxia arising from parasitised erythrocytes obstructing blood flow through vulnerable organs, particularly the brain, and we summarise why these notions are no longer tenable. In our view, this harmful sequestration is readily accommodated within the cytokine storm perspective as one of its secondary effects. We approach these issues by examining aspects of malaria, sepsis and influenza in parallel, and discuss the insights that comparisons of the literature can provide on the validity of possible anti-disease therapies.


Subject(s)
Cytokines/biosynthesis , Cytokines/physiology , Inflammation/immunology , Malaria/immunology , Humans , Inflammation/metabolism , Malaria/metabolism
3.
Malar J ; 5: 85, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-17029647

ABSTRACT

Malaria causes an acute systemic human disease that bears many similarities, both clinically and mechanistically, to those caused by bacteria, rickettsia, and viruses. Over the past few decades, a literature has emerged that argues for most of the pathology seen in all of these infectious diseases being explained by activation of the inflammatory system, with the balance between the pro and anti-inflammatory cytokines being tipped towards the onset of systemic inflammation. Although not often expressed in energy terms, there is, when reduced to biochemical essentials, wide agreement that infection with falciparum malaria is often fatal because mitochondria are unable to generate enough ATP to maintain normal cellular function. Most, however, would contend that this largely occurs because sequestered parasitized red cells prevent sufficient oxygen getting to where it is needed. This review considers the evidence that an equally or more important way ATP deficiency arises in malaria, as well as these other infectious diseases, is an inability of mitochondria, through the effects of inflammatory cytokines on their function, to utilise available oxygen. This activity of these cytokines, plus their capacity to control the pathways through which oxygen supply to mitochondria are restricted (particularly through directing sequestration and driving anaemia), combine to make falciparum malaria primarily an inflammatory cytokine-driven disease.


Subject(s)
Cytokines/metabolism , Inflammation/metabolism , Malaria/metabolism , Humans
4.
J Immunol ; 177(8): 5595-603, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015747

ABSTRACT

Increased arginase I activity is associated with allergic disorders such as asthma. How arginase I contributes to and is regulated by allergic inflammatory processes remains unknown. CD4+ Th2 lymphocytes (Th2 cells) and IL-13 are two crucial immune regulators that use STAT6-dependent pathways to induce allergic airways inflammation and enhanced airways responsiveness to spasmogens (airways hyperresponsiveness (AHR)). This pathway is also used to activate arginase I in isolated cells and in hepatic infection with helminths. In the present study, we show that arginase I expression is also regulated in the lung in a STAT6-dependent manner by Th2-induced allergic inflammation or by IL-13 alone. IL-13-induced expression of arginase I correlated directly with increased synthesis of urea and with reduced synthesis of NO. Expression of arginase I, but not eosinophilia or mucus hypersecretion, temporally correlated with the development, persistence, and resolution of IL-13-induced AHR. Pharmacological supplementation with l-arginine or with NO donors amplified or attenuated IL-13-induced AHR, respectively. Moreover, inducing loss of function of arginase I specifically in the lung by using RNA interference abrogated the development of IL-13-induced AHR. These data suggest an important role for metabolism of l-arginine by arginase I in the modulation of IL-13-induced AHR and identify a potential pathway distal to cytokine receptor interactions for the control of IL-13-mediated bronchoconstriction in asthma.


Subject(s)
Arginase/antagonists & inhibitors , Arginase/physiology , Bronchial Hyperreactivity/etiology , Interleukin-13/physiology , RNA Interference , Animals , Arginase/genetics , Arginase/metabolism , Arginine/metabolism , Asthma/etiology , Asthma/metabolism , Gene Expression Regulation, Enzymologic , Lung/enzymology , Lung/pathology , Mice , Mice, Knockout , Nitric Oxide Donors/metabolism , STAT6 Transcription Factor , Th2 Cells
5.
J Neuroimmunol ; 176(1-2): 1-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16730804

ABSTRACT

EAE development in SJL/J mice is age and sex dependent: young males are EAE resistant; females and adult males are EAE susceptible. By studying splenocytes' IFNgamma and NO production and the induction or the suppression of actively induced EAE by manipulating NO systemic levels, we provide evidence that the failure of young male SJL/J mice to develop EAE lies in the activation of the innate immune system by the immunising stimulus.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Nitric Oxide/physiology , Age Factors , Animals , Arginase/metabolism , Female , Immunity, Innate , Interferon-gamma/biosynthesis , Lymph Nodes/pathology , Male , Mice , Nitric Oxide Donors/pharmacology , Reactive Nitrogen Species/blood , Sex Characteristics , Spleen/pathology
6.
Am J Pathol ; 166(1): 147-57, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15632008

ABSTRACT

The Brown Norway (BN) rat is reported to be resistant to the induction of experimental autoimmune encephalomyelitis (EAE) and a number of mechanisms have been suggested to explain this resistance. In work reported here we provide evidence that such resistance in the BN rat can be accounted for, at least in part, by their ability to produce higher levels of nitric oxide (NO) than susceptible strains of rats. Spleen cells from the BN rat make significantly more NO following in vitro stimulation than do cells from the Lewis or PVG rat and following in vivo immunization using complete Freund's adjuvant (CFA) the BN rat makes substantially more NO than either susceptible strain. If carbonyl iron is used as adjuvant in vivo there is no increase in NO levels in the BN rat and they are rendered highly susceptible to EAE. Immunizing with CFA simultaneously with neuroantigen and carbonyl iron drives up NO levels and the resistance is restored. EAE produced using carbonyl iron is characterized by extensive macrophage/microglia presence in the central nervous system lesions of the BN rat yet the cytokine profile in the lymph nodes does not differ from that in the EAE Lewis rats.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Nitric Oxide/physiology , Animals , Base Sequence , Cytokines/genetics , DNA Primers , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunohistochemistry , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Polymerase Chain Reaction , Rats , Rats, Inbred BN , Rats, Inbred Lew
7.
Clin Exp Hypertens ; 26(6): 525-35, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15554455

ABSTRACT

(Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETA/NO) is a recently synthesized member of NO-releasing, polyamine zwitterions, the so-called NONOates, that spontaneously liberate NO in aqueous solutions. The aim of this study was to determine the hemodynamic effects of DETA/ NO in normotensive and hypertensive mice. Male Swiss Outbred mice were implanted with TA11PA-C20 blood pressure devices (Data Sciences International, USA). After recovery (7-10 days), blood pressure was monitored for 10 days while mice were receiving saline (0.1 ml/20 g/day, s.c.). Mice were then treated every four hours for 1 day with either DETA/NO 60 mg/kg i.p. or the inactive metabolite, diethylenetriamine 38 mg/kg (molar equivalent) i.p. After a 2 week wash-out period, mice were treated with adrenocorticotrophic hormone (ACTH: 500 microg/kg/day, s.c.) for 10 days and re-challenged with DETA/NO or diethylenetriamine. Results were expressed as mean +/- SEM. After 10 days of saline treatment, baseline systolic and diastolic blood pressure (BP) were similar for animals subsequently receiving DETA/NO or the amine (123 +/- 1/95 +/- 3 and 124 +/- 1/92 +/- 0.2 mmHg) respectively. DETA/NO induced a profound fall in BP [Systolic: 74 +/- 4 mmHg (-40 +/- 3%); Diastolic: 46 +/- 4 mmHg (-52 +/- 4%)] and an increase in heart rate [729 +/- 33 bpm (32 +/- 2%)] within the first 80 minutes. Diethylenetriamine had no effect. ACTH treatment increased BP in both groups (137 +/- 16/108 +/- 12 and 161 +/- 1/142 +/- 1 mmHg) respectively. DETA/ NO induced a profound fall in blood pressure [Systolic: 92 +/- 11 mmHg (-32 +/- 7%); Diastolic: 68 +/- 10 mmHg (-35 +/- 10%)] and an increase in heart rate [613 +/- 36 bpm (18 +/- 6%)] within the first 80 minutes. Again diethylenetriamine had no significant effect. There was no significant effect on body weight with any treatment. Thus DETA/NO has potent blood pressure lowering effects in both normotensive and hypertensive mice.


Subject(s)
Blood Pressure/drug effects , Hypertension/drug therapy , Nitric Oxide Donors/pharmacology , Triazenes/pharmacology , Adrenocorticotropic Hormone/pharmacology , Animals , Animals, Outbred Strains , Hypertension/chemically induced , Hypertension/physiopathology , Male , Mice , Sodium Chloride/pharmacology
8.
Clin Microbiol Rev ; 17(3): 509-39, table of contents, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15258091

ABSTRACT

There is now wide acceptance of the concept that the similarity between many acute infectious diseases, be they viral, bacterial, or parasitic in origin, is caused by the overproduction of inflammatory cytokines initiated when the organism interacts with the innate immune system. This is also true of certain noninfectious states, such as the tissue injury syndromes. This review discusses the historical origins of these ideas, which began with tumor necrosis factor (TNF) and spread from their origins in malaria research to other fields. As well the more established proinflammatory mediators, such as TNF, interleukin-1, and lymphotoxin, the roles of nitric oxide and carbon monoxide, which are chiefly inhibitory, are discussed. The established and potential roles of two more recently recognized contributors, overactivity of the enzyme poly(ADP-ribose) polymerase 1 (PARP-1) and the escape of high-mobility-group box 1 (HMGB1) protein from its normal location into the circulation, are also put in context. The pathogenesis of the disease caused by falciparum malaria is then considered in the light of what has been learned about the roles of these mediators in these other diseases, as well as in malaria itself.


Subject(s)
Inflammation/physiopathology , Malaria, Falciparum/physiopathology , Protozoan Infections/physiopathology , Sepsis/physiopathology , Virus Diseases/physiopathology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Humans , Inflammation/immunology , Malaria, Falciparum/immunology , Protozoan Infections/immunology , Sepsis/immunology , Virus Diseases/immunology
9.
Virology ; 326(1): 1-5, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15262489

ABSTRACT

Mice treated with a single injection of formalin-fixed Coxiella burnetii showed a significant increase in resistance to vaccinia virus (VV) infection compared to untreated mice. C. burnetii stimulated dramatically high levels of nitric oxide (NO) in the serum of treated mice, suggesting that NO might play a role in resistance to virus infection. To test this hypothesis, the effect of C. burnetii treatment on VV replication was examined in NOS2-/- and wild-type mice. C. burnetii treatment inhibited VV replication in both the knockout and wild-type mice but the effect was significantly greater in the NOS2-/- mice. Experiments in IFNgamma receptor knockout mice indicated that the nonspecific antiviral immunity induced by C. burnetii was dependent on IFNgamma and not NO. In the absence of NO, indoleamine 2,3-dioxygenase (IDO) was increased in C. burnetii-treated mice and this may contribute to the accelerated virus clearance in NOS2-/- mice.


Subject(s)
Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Coxiella burnetii/immunology , Nitric Oxide/blood , Vaccination , Vaccinia virus , Vaccinia/prevention & control , Animals , Antigens, Bacterial/immunology , Disease Models, Animal , Female , Formaldehyde , Indoleamine-Pyrrole 2,3,-Dioxygenase , Interferon-gamma/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Tryptophan Oxygenase/metabolism , Vaccinia/blood , Vaccinia/metabolism , Vaccinia virus/physiology , Virus Replication/immunology , Interferon gamma Receptor
10.
Pharmacol Ther ; 99(2): 221-60, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12888113

ABSTRACT

Falciparum malaria is a complex disease with no simple explanation, affecting organs where the parasite is rare as well as those organs where it is more common. We continue to argue that it can best be understood in terms of excessive stimulation of normally useful pathways mediated by inflammatory cytokines, the prototype being tumor necrosis factor (TNF). These pathways involve downstream mediators, such as nitric oxide (NO) that the host normally uses to control parasites, but which, when uncontrolled, have bioenergetic failure of patient tissues as their predictable end point. Falciparum malaria is no different from many other infectious diseases that are clinically confused with it. The sequestration of parasitized red blood cells, prominent in some tissues but absent in others with equal functional loss, exacerbates, but does not change, these overriding principles. Recent opportunities to stain a wide range of tissues from African pediatric cases of falciparum malaria and sepsis for the inducible NO synthase (iNOS) and migration inhibitory factor (MIF) have strengthened these arguments considerably. The recent demonstration of bioenergetic failure in tissue removed from sepsis patients being able to predict a fatal outcome fulfils a prediction of these principles, and it is plausible that this will be demonstrable in severe falciparum malaria. Understanding the disease caused by falciparum malaria at a molecular level requires an appreciation of the universality of poly(ADP-ribose) polymerase-1 (PARP-1) and Na(+)/K(+)-ATPase and the protean effects of activation by inflammation of the former that include inactivation of the latter.


Subject(s)
Cytokines/immunology , Hypoxia/complications , Immunity, Cellular/immunology , Inflammation/chemically induced , Malaria, Falciparum/physiopathology , Toxins, Biological/toxicity , Africa , Child , Cytokines/adverse effects , Cytokines/physiology , Humans , Inflammation/complications , Inflammation/physiopathology , Malaria, Falciparum/ethnology , Malaria, Falciparum/immunology , Mortality , Population Groups , Toxins, Biological/isolation & purification
11.
FASEB J ; 17(10): 1337-9, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12759332

ABSTRACT

Nitric oxide (NO) inhibits both actively induced and transferred autoimmune encephalomyelitis. To explore potential mechanisms, we examined the ability of NO to inhibit migration of T lymphoblasts through both collagen matrices and monolayers of rat brain endothelial cells. The NO donor 1-hydroxy-2-oxo-3, 3-bis (2-aminoethyl)-1-triazene (HOBAT) inhibited migration in a concentration-dependent manner. NO pretreatment of T cells inhibited migration through untreated endothelial cells, but NO pretreatment of endothelial cells had no inhibitory effect on untreated T cells. Therefore NO's migration inhibitory action was mediated through its effect on T cells and not endothelial cells. HOBAT did not inhibit migration by inducing T-cell death but rather by polarizing the T cells, resulting in a morphology suggestive of migrating cells. P70S6 kinase, shown to have a role in NO-induced migration inhibition in fibroblasts, had no role in the inhibitory effect of NO on T-cell migration. Thus, HOBAT did not alter p70S6K activity nor did rapamycin, a specific inhibitor of p70S6K, inhibit HOBAT-induced T-cell morphological changes or T-cell migration. We suggest that NO-induced morphological changes result in T cells with predefined migratory directionality, thus limiting the ability of these cells to respond to other migratory signals.


Subject(s)
Actin Cytoskeleton/ultrastructure , Actins/analysis , Cell Movement , Encephalomyelitis, Autoimmune, Experimental/immunology , Nitric Oxide/physiology , T-Lymphocytes/immunology , Animals , Brain/cytology , Cell Adhesion Molecules/metabolism , Cell Death , Cell Line , Cell Movement/drug effects , Cell Polarity , Cells, Cultured , Dogs , Endothelium/physiology , Lymphocyte Activation , Models, Immunological , Nitric Oxide Donors/pharmacology , Rats , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Stress Fibers/ultrastructure , T-Lymphocytes/cytology , T-Lymphocytes/ultrastructure , Tight Junctions/ultrastructure , Triazenes/pharmacology
12.
Circ Res ; 92(8): e70-7, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12690039

ABSTRACT

Percutaneous transluminal coronary angioplasty is a frequently used interventional technique to reopen arteries that have narrowed because of atherosclerosis. Restenosis, or renarrowing of the artery shortly after angioplasty, is a major limitation to the success of the procedure and is due mainly to smooth muscle cell accumulation in the artery wall at the site of balloon injury. In the present study, we demonstrate that the antiangiogenic sulfated oligosaccharide, PI-88, inhibits primary vascular smooth muscle cell proliferation and reduces intimal thickening 14 days after balloon angioplasty of rat and rabbit arteries. PI-88 reduced heparan sulfate content in the injured artery wall and prevented change in smooth muscle phenotype. However, the mechanism of PI-88 inhibition was not merely confined to the antiheparanase activity of this compound. PI-88 blocked extracellular signal-regulated kinase-1/2 (ERK1/2) activity within minutes of smooth muscle cell injury. It facilitated FGF-2 release from uninjured smooth muscle cells in vitro, and super-released FGF-2 after injury while inhibiting ERK1/2 activation. PI-88 inhibited the decrease in levels of FGF-2 protein in the rat artery wall within 8 minutes of injury. PI-88 also blocked injury-inducible ERK phosphorylation, without altering the clotting time in these animals. Optical biosensor studies revealed that PI-88 potently inhibited (Ki 10.3 nmol/L) the interaction of FGF-2 with heparan sulfate. These findings show for the first time the capacity of this sulfated oligosaccharide to directly bind FGF-2, block cellular signaling and proliferation in vitro, and inhibit injury-induced smooth muscle cell hyperplasia in two animal models. As such, this study demonstrates a new role for PI-88 as an inhibitor of intimal thickening after balloon angioplasty. The full text of this article is available online at http://www.circresaha.org.


Subject(s)
Angioplasty, Balloon/adverse effects , Muscle, Smooth, Vascular/drug effects , Oligosaccharides/pharmacology , Tunica Intima/drug effects , Animals , Binding, Competitive , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/pathology , Carotid Artery Injuries/prevention & control , Cell Division/drug effects , Enzyme Activation/drug effects , Fibroblast Growth Factor 2/metabolism , Heparitin Sulfate/metabolism , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Oligosaccharides/metabolism , Rabbits , Rats , Rats, Wistar , Signal Transduction/drug effects , Tunica Intima/metabolism , Tunica Intima/pathology , Tunica Media/drug effects , Tunica Media/metabolism , Tunica Media/pathology , Whole Blood Coagulation Time
13.
Xenotransplantation ; 9(3): 169-82, 2002 May.
Article in English | MEDLINE | ID: mdl-11983015

ABSTRACT

The rejection of pig proislet xenografts in mice is a CD4 T cell-dependent process in which macrophages play an important role. To assess the potential for activated macrophages to act as effector cells in xenograft destruction, we have examined the relationship between proislet xenograft rejection, two principal markers of macrophage activation, transcription of inducible nitric oxide synthase (iNOS) and production of nitric oxide (NO), and their temporal relationship to intragraft cytokine gene expression. Xenograft rejection in CBA/H mice correlated with early induction of intragraft host iNOS mRNA and marked intragraft production of NO (reactive nitrogen intermediates, RNI). Intragraft mRNA expression for IFN-gamma, IL-1beta and TNF, cytokines associated with macrophage activation, was also found. These findings suggested that activated macrophages could be contributing to xenograft destruction via local NO-mediated toxicity at the graft site. To test the role of NO in this model: (1) Q-fever antigen (QFA) was administered to recipient mice in order to induce high systemic RNI levels and (2) in another experiment, pig proislets were transplanted into iNOS-/- mice. Treatment with QFA correlated with prolonged xenograft survival at 7 days post-transplant. Splenocytes from QFA-treated, but not control mice at 7 and 22 days post-transplant, exhibited inhibition of secondary xenogeneic mouse antiporcine mixed lymphocyte reaction (MLR) that was reversed by culture with the NOS inhibitor N-methylarginine (NMA). Despite continued elevated NO production, xenograft protection was temporary with complete rejection by day 22. Evidence that locally produced NO was not contributing to rejection was seen when pig proislets transplanted into iNOS-/- mice were rejected with normal kinetics; in these animals intragraft NO production was not detected (despite porcine iNOS gene expression). Failure of activated macrophages to achieve indefinite xenograft survival suggests that other factors are also required. Macrophage potential to effect either destructive or protective roles after pig proislet xenotransplantation suggests that such functions may depend on the site and magnitude of macrophage activation. Together these findings clearly demonstrate that high NO levels in the periphery are not damaging to xenogeneic islet tissue, neither host nor donor NO production is essential for islet xenograft rejection and consequently elevated plasma RNI levels do not represent a direct marker for rejection.


Subject(s)
Graft Rejection/etiology , Graft Rejection/metabolism , Islets of Langerhans Transplantation/adverse effects , Nitric Oxide/metabolism , Animals , Cytokines/genetics , Fetal Tissue Transplantation/adverse effects , Fetal Tissue Transplantation/immunology , Fetal Tissue Transplantation/physiology , Graft Rejection/pathology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/physiology , Lymphocyte Activation , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , RNA, Messenger/genetics , Reactive Nitrogen Species/metabolism , Swine , T-Lymphocytes/immunology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...