Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Kidney Int ; 82(4): 412-27, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22495296

ABSTRACT

Endothelial progenitor cells are known to reverse acute kidney injury by paracrine mechanisms. We previously found that microvesicles released from these progenitor cells activate an angiogenic program in endothelial cells by horizontal mRNA transfer. Here, we tested whether these microvesicles prevent acute kidney injury in a rat model of ischemia-reperfusion injury. The RNA content of microvesicles was enriched in microRNAs (miRNAs) that modulate proliferation, angiogenesis, and apoptosis. After intravenous injection following ischemia-reperfusion, the microvesicles were localized within peritubular capillaries and tubular cells. This conferred functional and morphologic protection from acute kidney injury by enhanced tubular cell proliferation, reduced apoptosis, and leukocyte infiltration. Microvesicles also protected against progression of chronic kidney damage by inhibiting capillary rarefaction, glomerulosclerosis, and tubulointerstitial fibrosis. The renoprotective effect of microvesicles was lost after treatment with RNase, nonspecific miRNA depletion of microvesicles by Dicer knock-down in the progenitor cells, or depletion of pro-angiogenic miR-126 and miR-296 by transfection with specific miR-antagomirs. Thus, microvesicles derived from endothelial progenitor cells protect the kidney from ischemic acute injury by delivering their RNA content, the miRNA cargo of which contributes to reprogramming hypoxic resident renal cells to a regenerative program.


Subject(s)
Acute Kidney Injury/prevention & control , Cell-Derived Microparticles/transplantation , Endothelial Cells/transplantation , Kidney/metabolism , MicroRNAs/metabolism , Reperfusion Injury/prevention & control , Stem Cell Transplantation , Stem Cells , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Apoptosis , Capillaries/metabolism , Capillaries/pathology , Cell Hypoxia , Cell Proliferation , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Cells, Cultured , Chemotaxis, Leukocyte , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Gene Expression Regulation , Kidney/blood supply , Kidney/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Oligonucleotides/metabolism , RNA Interference , Rats , Rats, Wistar , Regeneration , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Ribonuclease III/genetics , Ribonuclease III/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Time Factors , Transfection
2.
Exp Neurol ; 234(1): 230-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22230666

ABSTRACT

Subarachnoid hemorrhage (SAH) is still a major cause of morbidity and mortality. α-Melanocyte stimulating hormone (α-MSH) and other melanocortin peptides exert potent neuroprotective action and they might modulate key molecules involved in SAH-induced vasospasm. The aim of this research was to determine whether treatment with the α-MSH analog Nle4,DPhe7-α-MSH (NDP-MSH) exerts protective effects in experimental SAH in the rat. Initial experiments examined effects of NDP-MSH on the basilar artery phenotype in the absence of injury. In these tests intrathecal injection of small concentrations (10ng) of the peptide induced a tolerant phenotype similar to that observed after ischemic preconditioning. Then the effect of systemic treatment with NDP-MSH (100µg i.v.) on experimental SAH was evaluated. SAH was induced by a single-blood injection into the cisterna magna. The basilar artery phenotype was examined at 4h and the artery caliber at 5days following SAH. Expression of 96 genes was analyzed by real-time reverse transcription polymerase chain reaction (RT-PCR) using Custom Taqman Low-Density Arrays. Four hours after SAH, the transcriptional profile of the basilar artery was deeply disrupted. Transcript alteration included genes involved in inflammation, stress response, apoptosis, and vascular remodeling. Treatment with NDP-MSH prevented most of these transcription changes and decreased phosphorylation of extracellular-signal-regulated kinases (ERK1/2) and inhibitor protein IκBα. Vasospasm on day 5 was significantly reduced by NDP-MSH administration. These results combine with others on CNS inflammation to suggest that the melanocortins could be safe and effective therapeutic candidates to treat SAH-related complications.


Subject(s)
Gene Expression Regulation/drug effects , Neuroprotective Agents/therapeutic use , Subarachnoid Hemorrhage/drug therapy , Vasospasm, Intracranial/prevention & control , alpha-MSH/analogs & derivatives , Analysis of Variance , Animals , Basilar Artery/drug effects , Basilar Artery/pathology , Disease Models, Animal , Gene Expression Profiling , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , MAP Kinase Signaling System/drug effects , Male , Oligonucleotide Array Sequence Analysis , RNA, Messenger , Random Allocation , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/complications , Vasospasm, Intracranial/etiology , alpha-MSH/therapeutic use
3.
Anesthesiology ; 116(3): 692-700, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22266570

ABSTRACT

BACKGROUND: Melanocortin peptides improve hemodynamic parameters and prevent death during severe hemorrhagic shock. In the present research we determined influences of a synthetic melanocortin 1/4 receptor agonist on the molecular changes that occur in rat liver during hemorrhage. METHODS: Controlled-volume hemorrhage was performed in adult rats under general anesthesia by a stepwise blood withdrawal until mean arterial pressure fell to 40 mmHg. Then rats received either saline or the synthetic melanocortin 1/4 receptor agonist Butir-His-D-Phe-Arg-Trp-Sar-NH2 (Ro27-3225; n = 6-8 per group). Hemogasanalysis was performed throughout a 60-min period. Gene expression in liver samples was determined at 1 or 3 h using quantitative real-time polymerase chain reaction. RESULTS: At 1 h, in saline-treated shocked rats, there were significant increases in activating transcription factor 3 (Atf3), early growth response 1 (Egr1), heme oxygenase (decycling) 1 (Hmox1), FBJ murine osteosarcoma viral oncogene homolog (Fos), and jun oncogene (Jun). These changes were prevented by Ro27-3225 (mean ± SEM: Atf3 152.83 ± 58.62 vs. 579.00 ± 124.13, P = 0.002; Egr1 13.21 ± 1.28 vs. 26.63 ± 1.02, P = 0.001; Hmox1 3.28 ± 0.31 vs. 166.54 ± 35.03, P = 0.002; Fos 4.36 ± 1.03 vs. 14.90 ± 3.44, P < 0.001; Jun 6.62 ± 1.93 vs. 15.07 ± 2.09, P = 0.005; respectively). Increases in alpha-2-macroglobulin (A2m), heat shock 70kD protein 1A (Hspa1a), erythropoietin (Epo), and interleukin-6 (Il6) occurred at 3 h in shocked rats and were prevented by Ro27-3225 treatment (A2m 6.90 ± 0.82 vs. 36.73 ± 4.00, P < 0.001; Hspa1a 10.34 ± 3.28 vs. 25.72 ± 3.64, P = 0.001; Epo 0.49 ± 0.13 vs. 2.37 ± 0.73, P = 0.002; Il6 1.05 ± 0.15 vs. 1.88 ± 0.23, P < 0.001; respectively). Further, at 3 h in shocked rats treated with Ro27-3225 there were significant increases in tight junction protein 1 (Tjp1; 27.30 ± 2.43 vs. 5.03 ± 1.68, P < 0.001) and nuclear receptor subfamily 4, group A, member 1 (Nr4a1; 91.03 ± 16.20 vs. 30.43 ± 11.0, P = 0.01) relative to sham animals. Treatment with Ro27-3225 rapidly restored blood pressure, hemogasanalysis parameters, and lactate blood levels. CONCLUSIONS: Melanocortin treatment significantly prevents most of the systemic and hepatic detrimental changes induced by hemorrhage.


Subject(s)
Melanocortins/therapeutic use , Peptides/therapeutic use , Shock, Hemorrhagic/drug therapy , Shock, Hemorrhagic/metabolism , Animals , Melanocortins/metabolism , Peptides/metabolism , Rats , Rats, Wistar , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/physiology , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/physiology , Shock, Hemorrhagic/genetics , Treatment Outcome
4.
Nephrol Dial Transplant ; 26(5): 1474-83, 2011 May.
Article in English | MEDLINE | ID: mdl-21324974

ABSTRACT

BACKGROUND: Several studies demonstrated that mesenchymal stem cells (MSCs) reverse acute kidney injury (AKI) by a paracrine mechanism rather than by MSC transdifferentiation. We recently demonstrated that microvesicles (MVs) released from MSCs may account for this paracrine mechanism by a horizontal transfer of messenger RNA and microRNA. METHODS: MVs isolated from MSCs were injected intravenously in rats (30 µg/rat) immediately after monolateral nephrectomy and renal artery and vein occlusion for 45 min. To evaluate the MV effects on AKI induced by ischaemia-reperfusion injury (IRI), the animals were divided into different groups: normal rats (n = 4), sham-operated rats (n = 6), IRI rats (n = 6), IRI + MV rats (n = 6), and IRI + RNase-MV rats (n = 6), and all animals were sacrificed at Day 2 after the operation. To evaluate the chronic kidney damage consequent to IRI, the rats were divided into different groups: sham-operated rats (n = 6) and IRI rats (n = 6), IRI + MV rats (n = 6), and all animal were sacrificed 6 months after the operation. RESULTS: We found that a single administration of MVs, immediately after IRI, protects rats from AKI by inhibiting apoptosis and stimulating tubular epithelial cell proliferation. The MVs also significantly reduced the impairment of renal function. Pretreatment of MVs with RNase to inactivate their RNA cargo abrogated these protective effects. Moreover, MVs by reducing the acute injury also protected from later chronic kidney disease. CONCLUSION: MVs released from MSCs protect from AKI induced by ischaemia reperfusion injury and from subsequent chronic renal damage. This suggest that MVs could be exploited as a potential new therapeutic approach.


Subject(s)
Acute Kidney Injury/prevention & control , Cell-Derived Microparticles , Kidney Failure, Chronic/prevention & control , Mesenchymal Stem Cells/physiology , Reperfusion Injury/physiopathology , Adult , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Kidney Tubules/cytology , Kidney Tubules/metabolism , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Rats , Rats, Sprague-Dawley
5.
ScientificWorldJournal ; 10: 1840-53, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20852827

ABSTRACT

Melanocortin peptides, the collective term for alpha-, beta-, and gamma-melanocyte-stimulating hormone (alpha-, beta-, gamma-MSH) and adrenocorticotropic hormone (ACTH), are elements of an ancient modulatory system. Natural melanocortins derive from the common precursor pro-opiomelanocortin (POMC). Five receptor subtypes for melanocortins (MC1-MC5) are widely distributed in brain regions and in peripheral cells. Melanocortin receptor activation by natural or synthetic ligands exerts marked anti-inflammatory and immunomodulatory effects. The anticytokine action and the inhibitory influences on inflammatory cell migration make melanocortins potential new drugs for treatment of inflammatory disorders. Effectiveness in treatment of acute, chronic, and systemic inflammatory disorders is well documented in preclinical studies. Further, melanocortins are promising compounds in neuroprotection. This review examines the main signaling circuits in anti-inflammatory and immunomodulatory actions of melanocortins, and the potential therapeutic use of these molecules.


Subject(s)
Brain/drug effects , Inflammation/prevention & control , Melanocortins/pharmacology , Receptors, Melanocortin/metabolism , Adrenocorticotropic Hormone/metabolism , Adrenocorticotropic Hormone/pharmacology , Animals , Brain/metabolism , Brain/pathology , Humans , Inflammation/metabolism , Melanocortins/metabolism , Models, Biological , Pro-Opiomelanocortin/metabolism , Signal Transduction/drug effects
6.
Adv Exp Med Biol ; 681: 117-25, 2010.
Article in English | MEDLINE | ID: mdl-21222264

ABSTRACT

Systemic inflammatory reactions are pivotal in many disorders and have important secondary influences in many more. Although inflammation is initially useful to limit infection, it can also be detrimental and cause organ failure. Modulation of systemic reactions is important to restrict mediator release and limit cell activation that could cause harmful consequences. Experiments in which different models and treatments were used show that melanocortins reduce host responses such as fever, shock, reperfusion injury and allograft rejection. Melanocortin-derived peptides could be an effective treatment to prevent organ failure caused by excessive production of pro-inflammatory mediators. The degree of the modulatory effect exerted by melanocortins should be sufficient to reduce severity of systemic inflammation without impairing the host defense mechanisms.


Subject(s)
Melanocortins/metabolism , Systemic Inflammatory Response Syndrome/metabolism , Systemic Inflammatory Response Syndrome/prevention & control , Animals , Humans
7.
Peptides ; 31(1): 116-22, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19799952

ABSTRACT

alpha-Melanocyte-stimulating hormone (alpha-MSH) is a pro-opiomelanocortin (POMC)-derived peptide that exerts multiple protective effects on host cells. Previous investigations showed that treatment with alpha-MSH or synthetic melanocortin agonists reduces heart damage in reperfusion injury and transplantation. The aim of this preclinical research was to determine whether melanocortin treatment induces preconditioning-like cardioprotection. In particular, the plan was to assess whether melanocortin administration causes phenotype changes similar to those induced by repetitive ischemic events. The idea was conceived because both ischemic preconditioning and melanocortin signaling largely depend on cAMP response element binding protein (CREB) phosphorylation. Rats received single i.v. injections of 750microg/kg of the alpha-MSH analogue Nle(4),DPhe(7)-alpha-MSH (NDP-MSH) or saline and were sacrificed at 0.5, 1, 3, or 5h. Western blot analysis showed that rat hearts expressed melanocortin 1 receptor (MC1R) protein. Treatment with NDP-MSH was associated with early and marked increase in interleukin 6 (IL-6) mRNA. This was followed by signal transducer and activator of transcription 3 (STAT3) phosphorylation and induction of suppressor of cytokine signaling 3 (SOCS3). There were no changes in expression of other cytokines of the IL-6 family. Expression of IL-10, IL-1beta, and TNF-alpha was likewise unaltered. In hearts of rats treated with NDP-MSH there was increased expression of the orphan nuclear receptor Nur77. The data indicate that NDP-MSH induces phenotype changes that closely resemble ischemic preconditioning and likely contribute to its established protection against reperfusion injury. In addition, the increased expression of Nur77 and SOCS3 could be part of a broader anti-inflammatory effect.


Subject(s)
Heart , Ischemic Preconditioning , Myocardium/metabolism , alpha-MSH/analogs & derivatives , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heart/anatomy & histology , Heart/drug effects , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Phenotype , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , alpha-MSH/pharmacology
8.
Brain Behav Immun ; 23(7): 877-84, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19394418

ABSTRACT

Acute brain injury and brain death exert detrimental effects on peripheral host cells. Brain-induced impairment of immune function makes patients more vulnerable to infections that are a major cause of morbidity and mortality after stroke, trauma, or subarachnoid hemorrhage (SAH). Systemic inflammation and organ dysfunction are other harmful consequences of CNS injury. Brain death, the most severe consequence of brain injury, causes inflammatory changes in peripheral organs that can contribute to the inferior outcome of organs transplanted from brain-dead donors. Understanding of the mechanisms underlying the detrimental effects of brain injury on peripheral organs remains incomplete. However, it appears that sympathetic nervous system (SNS)-activation contributes to elicit both inflammation and immunodepression. Indeed, norepinephrine (NE)-induced production of chemokines in liver and other organs likely participates in local and systemic inflammatory changes. Conversely, catecholamine-stimulated interleukin-10 (IL-10) production by blood monocytes exerts immunosuppressive effects. Activation of the hypothalamic-pituitary-adrenal axis (HPA) by increased inflammatory cytokines within the brain is a significant component in the CNS-induced immune function inhibition. Non-neurologic consequences of brain injury show impressive similarities regardless of the brain insult and appear to depend on altered neuroimmune circuits. Modulation of these circuits could reduce extra-brain damage and improve patient outcome in both vascular and traumatic brain injury.


Subject(s)
Brain Injuries/immunology , Brain Injuries/pathology , Neuroimmunomodulation/immunology , Animals , Humans , Hypothalamo-Hypophyseal System/immunology , Hypothalamo-Hypophyseal System/physiopathology , Organ Transplantation/pathology , Pituitary-Adrenal System/immunology , Pituitary-Adrenal System/physiopathology , Rats , Sympathetic Nervous System/immunology , Sympathetic Nervous System/physiopathology
9.
Brain Behav Immun ; 22(6): 817-23, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18178058

ABSTRACT

Prevention of graft dysfunction is a major objective in transplantation medicine. Previous research on experimental heart transplantation indicated that treatment with the immunomodulatory peptide alpha-melanocyte stimulating hormone (alpha-MSH) improves histopathology, prolongs allograft survival, and reduces expression of the main tissue injury mediators. Because calcium-handling is critical in heart graft function, we determined the effects of transplantation injury and influences of alpha-MSH treatment on representative calcium regulatory proteins in rat heart allografts. Hearts from Brown Norway rats were transplanted heterotopically into MHC incompatible Lewis rats. Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), protein kinase C epsilon (PKC epsilon), sarcoplasmic/endoplasmic reticulum calcium-ATPase 2 (SERCA2a), arrestin-beta1 (Arrb1), cholinergic receptor M2 (Chrm2), and inositol 1,4,5-triphosphate receptor 1 (InsP(3)R1) were examined in: (1) non-transplanted donor hearts; (2) allografts from saline-treated rats; and (3) allografts from rats treated with the synthetic alpha-MSH analog Nle4-DPhe7-alpha-MSH (NDP-alpha-MSH) (100 microg i.p. every 12h). Transplantation injury was associated with severe reduction in calcium regulatory protein transcription and expression level. NDP-alpha-MSH administration partly reversed inhibition of protein transcription and almost completely prevented protein loss. Finally, because certain effects of cyclic 3'-5'-adenosine monophosphate (cAMP) signaling on calcium handling in cardiac myocytes depend on activation of exchange protein directly activated by cAMP 1 (Epac1), we determined Epac1 mRNA and protein expression in heart allografts. Transplantation injury markedly reduced Epac1. NDP-alpha-MSH treatment significantly preserved both Epac1 protein and mRNA in the allografts. Administration of alpha-MSH or related melanocortins could reduce transplantation-induced dysfunction through protection of heart calcium regulatory proteins.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Heart Transplantation/methods , alpha-MSH/pharmacology , Animals , Arrestins/genetics , Arrestins/metabolism , Blotting, Western , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium-Binding Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP/genetics , Cyclic AMP/metabolism , Gene Expression/drug effects , Gene Expression Profiling , Graft Rejection/drug therapy , Graft Survival/drug effects , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Inositol 1,4,5-Trisphosphate Receptors , Male , Myocardium/metabolism , Myocardium/pathology , Protein Kinase C-epsilon/drug effects , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Rats , Rats, Inbred BN , Rats, Inbred Lew , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sarcoplasmic Reticulum Calcium-Transporting ATPases/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , beta-Arrestin 1 , beta-Arrestins
10.
Shock ; 27(3): 326-33, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17304115

ABSTRACT

Alpha-melanocyte-stimulating hormone (alpha-MSH) is a peptide with broad anti-inflammatory effects. The present research was designed to determine production and effects of alpha-MSH in acute bleomycin-induced lung injury in rats. Intratracheal bleomycin instillation induced alpha-MSH expression in lung infiltrating cells and a marked peptide increase in the circulation. In experiments on the therapeutic potential of alpha-MSH on lung injury, we determined influences of the synthetic alpha-MSH analogue [Nle4-dPhe7]-alpha-MSH (NDP-alpha-MSH) on pulmonary edema, circulating nitric oxide, and gene expression profile in lungs 8 and 24 h after bleomycin instillation. Three main gene categories, known to be involved in the development of acute lung injury, were explored: stress response, inflammation, and fluid homeostasis. Peptide treatment was associated with a significant reduction in interstitial edema, with a virtually normal wet/dry weight ratio. Several stress-related genes, which were either upregulated or reduced by bleomycin, were only marginally altered during NDP-alpha-MSH treatment. NDP-alpha-MSH prevented bleomycin-related transcriptional alterations in genes involved in lung fluid homeostasis, including upregulation of Na/K-transporting ATPase and epithelial sodium channels and downregulation of cystic fibrosis transmembrane conductance regulator. Bleomycin-induced expression of proinflammatory and profibrotic factors (interleukin 6, tumor necrosis factor-alpha, transforming growth factor-beta1, and inducible nitric oxide synthase) and chemokines (chemokine [C-C motif] ligand 2 and chemokine [C-C motif] ligand 5) was likewise significantly reduced by NDP-alpha-MSH. In conclusion, treatment with the alpha-MSH analogue NDP-alpha-MSH greatly improved the clinical and molecular picture of bleomycin-induced lung injury. Treatment with alpha-MSH-related agents can exert beneficial effects in acute lung injury.


Subject(s)
Lung Injury , alpha-MSH/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Bleomycin/pharmacology , Gene Expression Profiling , Immunohistochemistry , Inflammation , Lung/metabolism , Lung/pathology , Male , Models, Biological , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Wistar
11.
Transplantation ; 82(1): 69-79, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16861944

ABSTRACT

BACKGROUND: There is evidence that brain death causes changes in peripheral organs. Marked inflammation is found in organs collected during experimental brain death and clinical studies indicate that, despite genetic mismatch, organs obtained from living donors show improved survival over those from brain-dead donors. The aim of the present clinical research was to explore changes in the transcriptional profile of livers from brain-dead organ donors. METHODS: Using the cDNA macroarray technique, we compared gene expression in liver biopsies from 21 brain-dead organ donors and in normal liver tissue obtained during resection of benign focal lesions. RESULTS: Analysis of gene expression showed significant differences in the mRNA levels of 117 genes. There was reduced expression of 93 genes whereas expression of 24 genes was enhanced. Downregulated pathways included transcripts related to morphogenesis, blood coagulation, complement cascade, amine metabolism, lipid metabolism, nucleic acid metabolism, biodegradation of xenobiotics, signal transduction, and transcription. Conversely, there was induction of genes related to acute phase response, damage-related response, electron transport, and energy metabolism. CONCLUSIONS: The present research demonstrates major changes in the transcriptional profile of livers from brain-dead organ donors. The presence of both down- and upregulated gene families suggests that the alteration in transcriptional profile is not a consequence of death-associated organ failure, but rather, an active change in regulatory mechanisms.


Subject(s)
Brain Death/metabolism , Gene Expression Profiling , Liver Transplantation , Liver/metabolism , Living Donors , Adult , Aged , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Transcription, Genetic
12.
Neuroimmunomodulation ; 13(1): 51-5, 2006.
Article in English | MEDLINE | ID: mdl-16864968

ABSTRACT

OBJECTIVE: There is evidence that brain death has detrimental effects on peripheral organs. Clinical and experimental studies on organ donors showed marked inflammation in tissue samples of livers and kidneys collected during brain death. The inflammatory reaction is characterized by release of cytokines and inflammatory cell infiltration. Because melanocortins and their receptors are significant modulators of inflammation, we hypothesized that downregulation of melanocortin receptors during brain death could contribute to enhance inflammation. METHODS: Using real-time polymerase chain reaction (PCR) analysis, we determined expression of melanocortin receptors in liver biopsies obtained from brain-dead organ donors before cold ischemia and in normal liver tissue during resection of benign focal lesions of the liver. Tissue biopsies were also analyzed for expression of intercellular adhesion molecule-1 (ICAM-1), which has a central function in inflammatory cell migration. RESULTS: Expression of melanocortin-1 receptor (MC1R) mRNA was markedly reduced in liver samples obtained from brain-dead organ donors compared to hepatic tissue collected during resection of benign focal lesions of the liver. Conversely, expression of the adhesion molecule ICAM-1 was significantly increased in livers of brain-dead organ donors. CONCLUSIONS: Disruption of the endogenous anti-inflammatory circuit based on MC1R could contribute to tissue damage during brain death.


Subject(s)
Brain Death/immunology , Brain Death/physiopathology , Hepatitis/immunology , Liver/immunology , Receptor, Melanocortin, Type 1/genetics , alpha-MSH/immunology , Adult , Aged , Biopsy , Cell Adhesion/immunology , Cell Death/immunology , Down-Regulation/immunology , Female , Hepatitis/metabolism , Hepatitis/physiopathology , Humans , Intercellular Adhesion Molecule-1/metabolism , Liver/metabolism , Liver/physiopathology , Male , Middle Aged , RNA, Messenger/metabolism , Up-Regulation/immunology , alpha-MSH/metabolism
13.
J Surg Res ; 131(2): 209-14, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16413580

ABSTRACT

BACKGROUND: alpha-Melanocyte stimulating hormone (alpha-MSH) is an endogenous peptide that has remarkable anti-inflammatory and antimicrobial effects. These activities have been traced to the C-terminal tripeptide Lys-Pro-Val (KPV). A dimer composed of two KPV sequences connected with a Cys-Cys linker, (CKPV)2, is currently under clinical investigation for antimicrobial use. The present research was designed to evaluate effects of (CKPV)(2) on endotoxin-induced host reactions in vitro and in vivo. MATERIALS AND METHODS: Effects of (CKPV)2, KPV, and [Nle4-dPhe7]-alpha-MSH (NDP-alpha-MSH) on tumor necrosis factor alpha (TNF-alpha) production were determined: 1) in human peripheral blood mononuclear cells (PBMC) stimulated with lipopolysaccharide (LPS) in vitro, and 2) in rats injected with LPS i.v. and sacrificed at 1 h. In additional experiments, dialysis peritonitis was induced in rats by adding LPS to dialysis fluid. Net ultrafiltrate was calculated and concentrations of nitrite (NO2-) and TNF-alpha were measured in blood and peritoneal fluid at 7 h. RESULTS: (CKPV)2 inhibited TNF-alpha production by LPS-stimulated human PBMC. This small peptide was as effective as NDP-alpha-MSH and more potent than KPV. Similar effectiveness was observed in vivo: 1 h after LPS injection, the large increase in circulating TNF-alpha was markedly reduced by (CKPV)2 treatment. In LPS-induced peritonitis, (CKPV)2 restored net ultrafiltrate to control values and significantly inhibited concentrations of TNF-alpha and NO2- both in plasma and in dialysate. CONCLUSIONS: The remarkable capacity of (CKPV)2 to inhibit endotoxin-induced host reactions suggests that it may be useful in treatment of inflammatory disorders.


Subject(s)
Lipopolysaccharides/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , alpha-MSH/physiology , Animals , Cell Culture Techniques , Leukocytes, Mononuclear , Nitric Oxide/analysis , Peptide Fragments/physiology , Peritoneal Dialysis , Rats , Rats, Wistar , alpha-MSH/analogs & derivatives
14.
J Immunol ; 175(5): 3391-401, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16116233

ABSTRACT

Novel therapies are sought to increase efficiency and survival of transplanted organs. Previous research on experimental heart transplantation showed that treatment with the anti-inflammatory peptide alpha-melanocyte-stimulating hormone (alpha-MSH) prolongs allograft survival. The aim of the present research was to determine the molecular mechanism of this protective activity. Gene expression profile was examined in heart grafts removed on postoperative days 1 and 4 from rats treated with saline or the synthetic alpha-MSH analog Nle4DPhe7 (NDP)-alpha-MSH. On postoperative day 1, the peptide induced expression of cytoskeleton proteins, intracellular kinases, transcription regulators, metallopeptidases, and protease inhibitors. Conversely, NDP-alpha-MSH repressed immune, inflammatory, cell cycle, and protein turnover mediators. Later effects of alpha-MSH treatment included down-regulation of oxidative stress response and up-regulation of ion channels, calcium regulation proteins, phosphatidylinositol signaling system, and glycolipidic metabolism. NDP-alpha-MSH exerted its effects on both Ag-dependent and -independent injury. The results indicate that NDP-alpha-MSH preserves heart function through a broad effect on multiple pathways and suggest that the peptide could improve the outcome of organ transplantation in combination with immunosuppressive treatments.


Subject(s)
Gene Expression Profiling , Heart Transplantation , Myocardium/metabolism , alpha-MSH/analogs & derivatives , Animals , Calcium/metabolism , Male , Rats , Rats, Inbred BN , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Homologous , alpha-MSH/therapeutic use
15.
Appl Biochem Biotechnol ; 101(1): 15-29, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12008864

ABSTRACT

Xylitol production by Debaryomyces hansenii NRRL Y-7426 was performed on synthetic medium varying the initial xylose concentration between 50 and 300 g/L. The experimental results of these tests were used to investigate the effect of substrate level on xylose consumption by this yeast. Satisfactory values of product yield on substrate (0.74-0.83 g/g) as well as volumetric productivity (0.481-0.694 g/L x h) were obtained over a wide range of xylose levels (90-200 g/L), while a worsening of kinetic parameters took place at higher concentration, likely due to a substrate inhibition phenomenon. The metabolic behavior of D. hansenii was studied, under these conditions, through a carbon material balance to estimate the fractions of xylose consumed by the cell for different activities (xylitol production, biomass growth, and respiration) during the lag, exponential, and stationary phases.


Subject(s)
Saccharomycetales/metabolism , Xylitol/biosynthesis , Xylose/metabolism , Biomass , Carbon/analysis , Carbon/metabolism , Carbon Dioxide/analysis , Chromatography, High Pressure Liquid , Electron Transport/physiology , Fermentation/physiology , Kinetics , Saccharomycetales/growth & development , Substrate Specificity , Xylose/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...