Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Int J Mol Sci ; 24(24)2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38139026

RESUMO

Adipose-derived stem cells (ASCs) have been used as a therapeutic intervention for peripheral artery disease (PAD) in clinical trials. To further explore the therapeutic mechanism of these mesenchymal multipotent stromal/stem cells in PAD, this study was designed to test the effect of xenogeneic ASCs extracted from human adipose tissue on hypoxic endothelial cells (ECs) and terminal unfolded protein response (UPR) in vitro and in an atherosclerosis-prone apolipoprotein E-deficient mice (ApoE-/- mice) hindlimb ischemia model in vivo. ASCs were added to Cobalt (II) chloride-treated ECs; then, metabolic activity, cell migration, and tube formation were evaluated. Fluorescence-based sensors were used to assess dynamic changes in Ca2+ levels in the cytosolic- and endoplasmic reticulum (ER) as well as changes in reactive oxygen species. Western blotting was used to observe the UPR pathway. To simulate an acute-on-chronic model of PAD, ApoE-/- mice were subjected to a double ligation of the femoral artery (DLFA). An assessment of functional recovery after DFLA was conducted, as well as histology of gastrocnemius. Hypoxia caused ER stress in ECs, but ASCs reduced it, thereby promoting cell survival. Treatment with ASCs ameliorated the effects of ischemia on muscle tissue in the ApoE-/- mice hindlimb ischemia model. Animals showed less muscle necrosis, less inflammation, and lower levels of muscle enzymes after ASC injection. In vitro and in vivo results revealed that all ER stress sensors (BIP, ATF6, CHOP, and XBP1) were activated. We also observed that the expression of these proteins was reduced in the ASCs treatment group. ASCs effectively alleviated endothelial dysfunction under hypoxic conditions by strengthening ATF6 and initiating a transcriptional program to restore ER homeostasis. In general, our data suggest that ASCs may be a meaningful treatment option for patients with PAD who do not have traditional revascularization options.


Assuntos
Células Endoteliais , Células-Tronco Mesenquimais , Humanos , Animais , Camundongos , Células Endoteliais/metabolismo , Neovascularização Fisiológica/fisiologia , Tecido Adiposo/metabolismo , Células-Tronco Mesenquimais/metabolismo , Hipóxia/metabolismo , Resposta a Proteínas não Dobradas , Isquemia/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo
2.
Sci Rep ; 13(1): 16159, 2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37758738

RESUMO

Systemic inflammation affects the whole vasculature, yet whether arterial and venous endothelial cells differ in their abilities to mediate inflammation and to return to homeostasis after an inflammatory stimulus has not been addressed thoroughly. We assessed gene-expression profiles in isolated endothelial cells from human umbilical arteries (HUAEC) or veins (HUVEC) under basal conditions, after TNF-α stimulation and various time points after TNF-α removal to allow reinstatement of homeostasis. TNF-α regulates the expression of different sets of transcripts that are significantly changed only in HUAEC, only in HUVEC or changed in both. We identified three types of gene regulation, i.e. genes that were significantly regulated after 24 h of TNF-α stimulation but no longer when TNF-α was removed (homeostatic regulation), genes that maintained significantly regulated after TNF-α removal (not homeostatic regulation) and genes that were only significantly regulated when TNF-α was removed (post-regulation). HUAEC and HUVEC quantitatively differed in these types of gene regulation, with relatively more genes being post-regulated in HUAEC. In conclusion our data demonstrate that HUAEC and HUVEC respond intrinsically different to an inflammatory insult. Whether this holds true for all endothelial cells and its relevance for inflammatory insults in different organs during systemic inflammation warrants further studies.


Assuntos
Células Endoteliais , Fator de Necrose Tumoral alfa , Humanos , Células Endoteliais/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Células Cultivadas , Regulação da Expressão Gênica , Inflamação/genética , Inflamação/metabolismo , Veias Umbilicais , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo
3.
Int J Mol Sci ; 24(15)2023 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-37569572

RESUMO

Innate immune memory allows macrophages to adequately respond to pathogens to which they have been pre-exposed. To what extent different pattern recognition receptors, cytokines and resolution signals influence innate immune memory needs further elucidation. The present study assessed whether lipopolysaccharide (LPS) tolerance in monocytes and macrophages is affected by these factors. Human CD14+ cells were isolated from peripheral blood, stimulated by LPS and re-stimulated after 3 days of resting. Hereafter, immune-responsive gene 1 (IRG-1), heme oxygenase 1 (HO-1), tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6) expression were assessed. Our study revealed the following findings: (1) While pre-stimulation with the Toll-like receptor 4 ligand LPS inhibits the induction of IRG-1, TNF-α and IL-6 expression, pre-stimulation with TLR 1/2 ligands only affects cytokine production but not IRG-1 expression upon subsequent TLR4 engagement. (2) Prior TNF-α stimulation does not affect LPS tolerance but rather increases LPS-mediated cytokine expression. (3) Dimethyl itaconate (DMI) inhibits the expression of IRG-1 in a dose-dependent manner but does not affect TNF-α or IL-6 expression. (4) Docosahexaenoic acid (DHA) partly inhibits IRG-1 expression in monocytes but not in M(IFNγ) and M(IL-4) polarized macrophages. LPS tolerance is not affected in these cells by DHA. The data presented in this study partly corroborate and extend previous findings on innate immune memory and warrant further studies on LPS tolerance to gain a better understanding of innate immune memory at the molecular level.


Assuntos
Lipopolissacarídeos , Monócitos , Humanos , Monócitos/metabolismo , Interleucina-6/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Macrófagos/metabolismo , Citocinas/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Tolerância Imunológica
4.
J Leukoc Biol ; 112(3): 437-447, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35075676

RESUMO

In atherosclerotic lesions, macrophages are exposed to CSFs and various microenvironmental cues, which ultimately drive their polarization state. We studied the expression of different CSFs in artery specimen and cultured vascular cells and assessed whether concurrent stimulation (CS) of monocytes with CSF1 and polarizing cytokines generated macrophages (CSM1 and CSM2) that were phenotypically and functionally different from classically polarized M1 and M2 macrophages. We also assessed the influence of acetylsalicylic acid (ASA) on the capacity of polarized macrophages to stimulate T-cell proliferation. CSF1 was the most prominent CSF expressed in arteries and cultured vascular cells. M1 and CSM1 macrophages differed in CD86 and CD14 expression, which was up-regulated respectively down-regulated by LPS. M2 and CSM2 macrophages were phenotypically similar. Cyclooxygenase expression was different in CSM1 (COX-1- and COX-2+ after LPS stimulation) and CSM2 (COX-1+ and COX-2- ) macrophages. TNFα production was more pronounced in CSM1 macrophages, whereas IL-10 was produced at higher levels by CSM2 macrophages. Proliferation of allogeneic T cells was strongly supported by CSM2, but not by CSM1 polarized macrophages. Although ASA did not affect anti-CD3/CD28-mediated proliferation, it significantly reduced CSM2 and CSM1-mediated T-cell proliferation. Supernatants of LPS-stimulated CSM2 but not of CSM1 macrophages could overcome the inhibition by ASA. Hence, we demonstrate that CSM1 and CSM2 macrophages are phenotypically and to some extent functionally distinct from classically polarized M1 and M2 macrophages. CSM2 macrophages produce a COX-1-dependent soluble factor that supports T-cell proliferation, the identity hereof is still elusive and warrants further studies.


Assuntos
Citocinas , Monócitos , Diferenciação Celular , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Monócitos/metabolismo
5.
Sci Rep ; 11(1): 8004, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33850227

RESUMO

Methylglyoxal (MGO), a precursor of advanced glycation end products (AGEs), is regarded as a pivotal mediator of vascular damage in patients with diabetes. We have previously reported that MGO induces transcriptional changes compatible with p53 activation in cultured human endothelial cells. To further substantiate this finding and to explore the underlying mechanisms and possible consequences of p53 activation, we aimed (1) to provide direct evidence for p53 activation in MGO-treated human umbilical vein endothelial cells (HUVECs), (2) to assess putative mechanisms by which this occurs, (3) to analyze down-stream effects on mTOR and autophagy pathways, and (4) to assess the potential benefit of carnosine herein. Exposure of HUVECs to 800 µM of MGO for 5 h induced p53 phosphorylation. This was paralleled by an increase in TUNEL and γ-H2AX positive cells, indicative for DNA damage. Compatible with p53 activation, MGO treatment resulted in cell cycle arrest, inhibition of mTORC1 and induction of autophagy. Carnosine co-treatment did not counteract MGO-driven effects. In conclusion, our results demonstrate that MGO elicits DNA damage and p53 activation in HUVECs, resulting in modulation of downstream pathways, e.g. mTORC1.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Aldeído Pirúvico/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Autofagia/efeitos dos fármacos , Carnosina/farmacologia , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Glicosilação/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
6.
Sci Rep ; 9(1): 1152, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30718683

RESUMO

Although methylglyoxal (MGO) has emerged as key mediator of diabetic microvascular complications, the influence of MGO on the vascular transcriptome has not thoroughly been assessed. Since diabetes is associated with low grade inflammation causing sustained nuclear factor-kappa B (NF-κB) activation, the current study addressed 1) to what extent MGO changes the transcriptome of human umbilical vein endothelial cells (HUVECs) exposed to an inflammatory milieu, 2) what are the dominant pathways by which these changes occur and 3) to what extent is this affected by carnosine, a putative scavenger of MGO. Microarray analysis revealed that exposure of HUVECs to high MGO concentrations significantly changes gene expression, characterized by prominent down-regulation of cell cycle associated genes and up-regulation of heme oxygenase-1 (HO-1). KEGG-based pathway analysis identified six significantly enriched pathways of which the p53 pathway was the most affected. No significant enrichment of inflammatory pathways was found, yet, MGO did inhibit VCAM-1 expression in Western blot analysis. Carnosine significantly counteracted MGO-mediated changes in a subset of differentially expressed genes. Collectively, our results suggest that MGO initiates distinct transcriptional changes in cell cycle/apoptosis genes, which may explain MGO toxicity at high concentrations. MGO did not augment TNF-α induced inflammation.


Assuntos
Ciclo Celular/efeitos dos fármacos , Genes cdc/efeitos dos fármacos , Aldeído Pirúvico/farmacologia , Carnosina/farmacologia , Heme Oxigenase-1/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
7.
Amino Acids ; 51(1): 17-25, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29961141

RESUMO

Low serum carnosinase (CN-1) concentrations are associated with low risk for development of diabetic nephropathy (DN) in patients with type 2 diabetes (T2D). Although CN-1 is expressed in the kidney, urinary CN-1 (CNU) excretion and its pathological relevance in patients with T2D have not been investigated to date. The present study therefore assessed the extent of CNU excretion in healthy subjects (n = 243) and in patients with T2D (n = 361) enrolled in the DIAbetes and LifEstyle Cohort Twente-1 (DIALECT-1) in relation to functional renal parameters. CNU was detected in a high proportion of healthy individuals, 180 (74%); median CNU excretion was 0.25 mg/24 h [(IQR 0-0.65 mg/24 h]. In patients with T2D the prevalence and extent of CNU increased in parallel with albuminuria (r = 0.59, p < 0.0001; median CNU 0.1 vs 0.2 vs 1.5 mg/24 h, p < 0.0001; prevalence of CNU 61 vs. 81 vs. 97% p < 0.05 in normo- (n = 241), micro- (n = 80) and macroalbuminuria (n = 40), respectively). Patients with estimated glomerular filtration rate (eGFR) < 30 ml/min/1.73 m2 displayed higher median CNU excretion rates in comparison to patients with preserved eGFR (> 90 ml/min/1.73 m2) (1.36 vs 0.13 mg/24 h, p < 0.05). Backward stepwise multivariate linear regression analysis revealed albuminuria, eGFR and glycosuria to be independent factors of CNU excretion rates, all together explaining 37% of variation of CNU excretion rates (R2 = 0.37, p < 0.0001). These results show for the first time that CN-1 can be detected in urine and warrants prospective studies to assess the relevance of CNU for renal function deterioration in diabetes patients.


Assuntos
Albuminúria/urina , Diabetes Mellitus Tipo 2/urina , Dipeptidases/urina , Rim/fisiopatologia , Idoso , Idoso de 80 Anos ou mais , Animais , Estudos de Coortes , Diabetes Mellitus Tipo 2/complicações , Feminino , Voluntários Saudáveis , Humanos , Modelos Lineares , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade
8.
Liver Transpl ; 24(10): 1336-1345, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30102825

RESUMO

Treatment of donation after brain death (DBD) donors with low-dose dopamine improves the outcomes after kidney and heart transplantation. This study investigates the course of liver allografts from multiorgan donors enrolled in the randomized dopamine trial between 2004 and 2007 (clinicaltrials.gov identifier: NCT00115115). There were 264 hemodynamically stable DBDs who were randomly assigned to receive low-dose dopamine. Dopamine was infused at 4 µg/kg/minute for a median duration of 6.0 hours (interquartile range, 4.4-7.5 hours). We assessed the outcomes of 212 liver transplantations (LTs) performed at 32 European centers. Donors and recipients of both groups were very similar in baseline characteristics. Pretransplant laboratory Model for End-Stage Liver Disease score was not different in recipients of a dopamine-treated versus untreated graft (18 ± 8 versus 20 ± 8; P = 0.12). Mean cold ischemia time was 10.6 ± 2.9 versus 10.1 ± 2.8 hours (P = 0.24). No differences occurred in biopsy-proven rejection episodes (14.4% versus 15.7%; P = 0.85), requirement of hemofiltration (27.9% versus 31.5%; P = 0.65), the need for early retransplantation (5.8% versus 6.5%; P > 0.99), the incidence of primary nonfunction (7.7% versus 8.3%; P > 0.99), and in-hospital mortality (15.4% versus 14.8%; P > 0.99). Graft survival was 71.2% versus 73.2% and 59.6% versus 62.0% at 2 and 3 years (log-rank P = 0.71). Patient survival was 76.0% versus 78.7% and 65.4% versus 69.4% at 1 and 3 years (log-rank P = 0.50). In conclusion, donor pretreatment with dopamine has no short-term or longterm effects on outcome after LT. Therefore, low-dose dopamine pretreatment can safely be implemented as the standard of care in hemodynamically stable DBDs.


Assuntos
Dopamina/administração & dosagem , Doença Hepática Terminal/cirurgia , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Fígado/efeitos adversos , Coleta de Tecidos e Órgãos/métodos , Adulto , Isquemia Fria/efeitos adversos , Doença Hepática Terminal/diagnóstico , Feminino , Rejeição de Enxerto/prevenção & controle , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Índice de Gravidade de Doença , Doadores de Tecidos , Resultado do Tratamento
9.
Clin Exp Rheumatol ; 36 Suppl 111(2): 143-151, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29745882

RESUMO

Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a potentially lethal autoimmune disease whose pathology comprises disturbed T cell differentiation and functionality accompanied by dysfunctional autoreactive immunoglobulin development, culminating in destructive innate immune response as well. Purines, adenine nucleotides and adenosine in particular, have been elucidated as potent extracellular mediators for fine adjustment of these pivotal processes establishing human immunity. Therefore, the extracellular purinergic microenvironment is under control of ectonucleotidases CD39 and CD73 degrading pro-inflammatory adenosine triphosphate (ATP) to anti-inflammatory adenosine as well as adenosine deaminase bound to CD26 deactivating adenosine. Accordingly, the ATP P2X7 receptor was elicited to be responsible for promotion of inflammation, while predominantly the adenosine A2A receptor demonstrated the opposite. Recent reports pointed at the adenosinergic system to be crucially involved in AAV pathogenesis. Here, experimental evidence on ecto-enzymes controlling extracellular adenine nucleotide concentrations and purinergic signaling in the immune system with respect to its contribution to the AAV pathomechanism is reviewed besides unsolved problems being identified that require further investigation in order to develop new treatment strategies for AAV.


Assuntos
Adenosina/imunologia , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/imunologia , Receptor A2A de Adenosina/imunologia , Receptores Purinérgicos P2X7/imunologia , 5'-Nucleotidase/metabolismo , Adenosina/metabolismo , Adenosina Desaminase/metabolismo , Trifosfato de Adenosina/metabolismo , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/metabolismo , Antígenos CD/metabolismo , Apirase/metabolismo , Diferenciação Celular , Humanos , Inflamação , Transdução de Sinais , Linfócitos T/imunologia
10.
Cell Physiol Biochem ; 46(2): 713-726, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29621776

RESUMO

BACKGROUND/AIMS: Reactive dicarbonyl compounds, such as methylglyoxal (MG), contribute to diabetic complications. MG-scavenging capacities of carnosine and anserine, which have been shown to mitigate diabetic nephropathy, were evaluated in vitro and in vivo. METHODS: MG-induced cell toxicity was characterized by MTT and MG-H1-formation, scavenging abilities by Western Blot and NMR spectroscopies, cellular carnosine transport by qPCR and microplate luminescence and carnosine concentration by HPLC. RESULTS: In vitro, carnosine and anserine dose-dependently reduced N-carboxyethyl lysine (CEL) and advanced glycation end products (AGEs) formation. NMR studies revealed the formation of oligo/polymeric products of MG catalyzed by carnosine or anserine. MG toxicity (0.3-1 mM) was dose-dependent for podocytes, tubular and mesangial cells whereas low MG levels (0.2 mM) resulted in increased cell viability in podocytes (143±13%, p<0.001) and tubular cells (129±3%, p<0.001). Incubation with carnosine/anserine did not reduce MG-induced toxicity, independent of incubation times and across large ranges of MG to carnosine/anserine ratios. Cellular carnosine uptake was low (<0.1% in 20 hours) and cellular carnosine concentrations remained unaffected. The putative carnosine transporter PHT1 along with the taurine transporter (TauT) was expressed in all cell types while PEPT1, PEPT2 and PHT2, also belonging to the proton-coupled oligopeptide transporter (POT) family, were only expressed in tubular cells. CONCLUSION: While carnosine and anserine catalyze the formation of MG oligo/polymers, the molar ratios required for protection from MG-induced cellular toxicity are not achievable in renal cells. The effect of carnosine in vivo, to mitigate diabetic nephropathy may therefore be independent upon its ability to scavenge MG and/or carnosine is mainly acting extracellularly.


Assuntos
Carnosina/química , Carnosina/metabolismo , Polímeros/química , Aldeído Pirúvico/química , Animais , Anserina/análise , Anserina/química , Anserina/metabolismo , Carnosina/análise , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Glutationa/análise , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Produtos Finais de Glicação Avançada/química , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Transportador 1 de Peptídeos/genética , Transportador 1 de Peptídeos/metabolismo , Podócitos/citologia , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Polímeros/metabolismo , Aldeído Pirúvico/toxicidade , Albumina Sérica/química , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Simportadores/genética , Simportadores/metabolismo
11.
PLoS One ; 13(3): e0195064, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29601599

RESUMO

CD34+/CD133+- cells are a bone marrow derived stem cell population, which presumably contain vascular progenitor cells and are associated with improved vascular repair. In this study, we investigated whether the adhesion molecules ICAM-1 (intercellular adhesion molecule-1), VCAM-1 (vascular adhesion molecule-1), E-selectin und L-selectin, which are involved in homing of vascular stem cells, are upregulated by CD34+/CD133+-stem cells from septic patients and would be associated with improved clinical outcome. Peripheral blood mononuclear cells from intensive care unit (ICU) patients with (n = 30) and without sepsis (n = 10), and healthy volunteers (n = 15) were isolated using Ficoll density gradient centrifugation. The expression of VCAM-1, ICAM-1, E-selectin and L-selectin was detected on CD34+/CD133+-stem cells by flow cytometry. The severity of disease was assessed by the Simplified Acute Physiology Score (SAPS) II. Serum concentrations of vascular endothelial growth factor (VEGF) and angiopoietin (Ang)-2 were determined by Enzyme-linked immunosorbent assay. The expression of VCAM-1, ICAM-1, E-selectin and L-selectin by CD34+/CD133+-stem cells was significantly upregulated in septic patients, and correlated with sepsis severity. Furthermore, high expression of VCAM-1 by CD34+/CD133+-stem cells revealed a positive association with mortalitiy (p<0.05). Furthermore, significantly higher serum concentrations of VEGF and Ang-2 were found in septic patients, however none showed a strong association with survival. Our data suggest, that VCAM-1 upregulation on CD34+/CD133+-stem cells could play a crucial role in their homing in the course of sepsis. An increase in sepsis severity resulted in both and increase in CD34+/CD133+-stem cells and VCAM-1-expression by those cells, which might reflect an increase in need for vascular repair.


Assuntos
Antígeno AC133/metabolismo , Antígenos CD34/metabolismo , Sepse/metabolismo , Sepse/patologia , Células-Tronco/metabolismo , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/metabolismo , Adulto , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Sepse/sangue , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/sangue , Proteínas de Transporte Vesicular/sangue
12.
Sci Rep ; 7(1): 11683, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28916770

RESUMO

Extracellular adenosine, generated via the concerted action of CD39 and CD73, contributes to T-cell differentiation and function. Adenosine concentrations are furthermore influenced by adenosine deaminase binding protein CD26. Because aberrant T-cell phenotypes had been reported in anti-neutrophil cytoplasmic auto-antibody (ANCA)-associated vasculitis (AAV) patients, an impaired expression of these molecules on T-cells of AAV patients was hypothesized in the present study. While in AAV patients (n = 29) CD26 was increased on CD4+ lymphocytes, CD39 and CD73 were generally reduced on patients' T-cells. In CD4+ cells significant differences in CD73 expression were confined to memory CD45RA- cells, while in CD4- lymphocytes differences were significant in both naïve CD45RA+ and memory CD45RA- cells. The percentage of CD4-CD73+ cells correlated with micro-RNA (miR)-31 expression, a putative regulator of factor inhibiting hypoxia-inducible factor 1 alpha (FIH-1), inversely with serum C-reactive protein (CRP) and positively with estimated glomerular filtration rate (eGFR). No correlation with disease activity, duration, and ANCA profile was found. It remains to be assessed if a decreased CD73 and CD39 expression underlies functional impairment of lymphocytes in AAV patients. Likewise, the relations between frequencies of CD4-CD73+ cells and serum CRP or eGFR require further functional elucidation.


Assuntos
Adenosina/metabolismo , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/patologia , Apirase/análise , Dipeptidil Peptidase 4/análise , Linfócitos T/patologia , 5'-Nucleotidase , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteína C-Reativa/análise , Feminino , Proteínas Ligadas por GPI , Taxa de Filtração Glomerular , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
13.
J Diabetes Res ; 2017: 9506730, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28553654

RESUMO

Considering that the homozygous CNDP1 (CTG)5 genotype affords protection against diabetic nephropathy (DN) in female patients with type 2 diabetes, this study assessed if this association remains gender-specific when applying clinical inclusion criteria (CIC-DN) or biopsy proof (BP-DN). Additionally, it assessed if the prevalence of the protective genotype changes with diabetes duration and time on hemodialysis and if this occurs in association with serum carnosinase (CN-1) activity. Whereas the distribution of the (CTG)5 homozygous genotype in the no-DN and CIC-DN patients was comparable, a lower frequency was found in the BP-DN patients, particularly in females. We observed a significant trend towards high frequencies of the (CTG)5 homozygous genotype with increased time on dialysis. This was also observed for diabetes duration but only reached significance when both (CTG)5 homo- and heterozygous patients were included. CN-1 activity negatively correlated with time on hemodialysis and was lower in (CTG)5 homozygous patients. The latter remained significant in female subjects after gender stratification. We confirm the association between the CNDP1 genotype and DN to be likely gender-specific. Although our data also suggest that (CTG)5 homozygous patients may have a survival advantage on dialysis and in diabetes, this hypothesis needs to be confirmed in a prospective cohort study.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/genética , Dipeptidases/genética , Falência Renal Crônica/genética , Idoso , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/terapia , Dipeptidases/sangue , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Falência Renal Crônica/etiologia , Falência Renal Crônica/patologia , Falência Renal Crônica/terapia , Masculino , Pessoa de Meia-Idade , Fenótipo , Polimorfismo Genético , Diálise Renal , Estudos Retrospectivos , Fatores de Tempo
14.
Exp Clin Transplant ; 14(3): 299-306, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27221721

RESUMO

OBJECTIVES: Retrospective studies suggest that donor desmopressin (DDAVP) treatment improves renal transplant outcome. The present study tests the hypothesis that desmopressin neutralizes the graft's endothelium from proinflammatory angiopoietin 2 containing Weibel-Palade bodies in the donor, resulting in reduced Weibel-Palade body release at the time of reperfusion in the recipient. MATERIALS AND METHODS: Using rat models, we examined the influence of desmopressin treatment on the expression of vasopressin 2 receptors and adhesion molecules in brain-dead donors, with renal function examined in allogeneic recipients. The influence of desmopressin on the expression of adhesion molecules also was tested in vitro. RESULTS: Vasopressin 2 receptors were restricted to collecting ducts and distal tubules and only scarcely found in the renal vasculature. Vasopressin 2 receptor expression was down-regulated in brain-dead rats by desmopressin. Renal expression of vascular cellular adhesion molecule 1 and intercellular adhesion molecule 1 were significantly reduced in these rats. In contrast, angiopoietin 2 did not influence the expression of adhesion molecules in in vitro cultured endothelial cells after tumor necrosis factor ? stimulation. Donor desmopressin treatment improved neither renal function nor histology in allogeneic renal transplant recipients. CONCLUSIONS: Our data do not support the hypothesis that the clinically observed salutary effect of desmopressin is mediated by depletion of Weibel-Palade bodies in renal allografts.


Assuntos
Desamino Arginina Vasopressina/farmacologia , Células Endoteliais/efeitos dos fármacos , Transplante de Rim/efeitos adversos , Rim/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Corpos de Weibel-Palade/efeitos dos fármacos , Angiopoietina-2/farmacologia , Animais , Células Cultivadas , Isquemia Fria/efeitos adversos , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Rim/metabolismo , Rim/patologia , Masculino , Modelos Animais , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Receptores de Vasopressinas/agonistas , Receptores de Vasopressinas/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Fatores de Tempo , Transplante Homólogo , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/metabolismo , Isquemia Quente/efeitos adversos , Corpos de Weibel-Palade/metabolismo , Corpos de Weibel-Palade/patologia
15.
J Diabetes Res ; 2016: 8710432, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26788523

RESUMO

Iron has been suggested to affect the clinical course of type 2 diabetes (T2DM) as accompanying increased intracellular iron accumulation may provide an alternative source for reactive oxygen species (ROS). Although carnosine has proven its therapeutic efficacy in rodent models of T2DM, little is known about its efficacy to protect cells from iron toxicity. We sought to assess if high glucose (HG) exposure makes cultured human umbilical vein endothelial cells (HUVECs) and renal proximal tubular epithelial cells (PTECs) more susceptible to metal induced toxicity and if this is ameliorated by L-carnosine. HUVECs and PTECs, cultured under normal glucose (5 mM, NG) or HG (30 mM), were challenged for 24 h with FeCl3. Cell viability was not impaired under HG conditions nor did HG increase susceptibility to FeCl3. HG did not change the expression of divalent metal transporter 1 (DMT1), ferroportin (IREG), and transferrin receptor protein 1 (TFRC). Irrespective of glucose concentrations L-carnosine prevented toxicity in a dose-dependent manner, only if it was present during the FeCl3 challenge. Hence our study indicates that iron induced cytotoxicity is not enhanced under HG conditions. L-Carnosine displayed a strong protective effect, most likely by chelation of iron mediated toxicity.


Assuntos
Carnitina/farmacologia , Cloretos/toxicidade , Células Epiteliais/efeitos dos fármacos , Compostos Férricos/toxicidade , Glucose/toxicidade , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Hiperglicemia/metabolismo , Quelantes de Ferro/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Antígenos CD/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citoproteção , Dipeptidases/genética , Dipeptidases/metabolismo , Relação Dose-Resposta a Droga , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Hiperglicemia/patologia , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Receptores da Transferrina/metabolismo , Transfecção
16.
Transplantation ; 100(1): 80-90, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26674731

RESUMO

BACKGROUND: Transplant vasculopathy (TV) is a major cause for late graft loss after cardiac transplantation. Endothelial damage and T cell infiltration play a pivotal role in the development of TV. Because N-octanoyl dopamine (NOD) inhibits vascular inflammation and suppresses T cell activation in vitro, we here tested the hypothesis that NOD treatment ameliorates TV. METHODS: Aortic grafts were orthotopically transplanted in the Dark Agouti to Brown Norway strain combination. Recipient rats were treated with NOD or vehicle administered via osmotic minipumps. Histology and quantitative polymerase chain reaction (qPCR) were performed on nontransplanted aortas and grafts explanted 2 and 4 weeks after transplantation to assess the degree of TV, inflammation, apoptosis, and number of (proliferating) α smooth muscle actin (αSMA) neointimal cells. In vitro analyses of human aortic smooth muscle cells were performed to test the effect of NOD on proliferation (WST-1 assay), cell cycle (flow cytometry and qPCR), and cytokine-induced apoptosis (flow cytometry). RESULTS: Allografts from vehicle-treated recipients developed neointimal lesions predominantly consisting of αSMA-expressing cells. NOD treatment significantly reduced neointima formation and neointimal αSMA cells. In situ, smooth muscle cell proliferation (Ki67) was not influenced by NOD. Macrophage (CD68), T (CD3), and Natural Killer (ANK61) cell infiltration as well as intragraft TNFα and IFNγ mRNA expression were similar in both groups. Medial apoptosis (cleaved caspase-3) was significantly reduced by NOD. In vitro, NOD inhibited proliferation of human aortic smooth muscle cells by causing a G1-arrest and protected from TNFα-induced apoptosis. CONCLUSIONS: This study identified NOD as potential treatment modality to attenuate TV. Our data clearly support a vasculoprotective effect of NOD by reducing smooth muscle cell proliferation and inflammation-induced apoptosis.


Assuntos
Anti-Inflamatórios/farmacologia , Doenças da Aorta/prevenção & controle , Dopamina/análogos & derivados , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/transplante , Actinas/metabolismo , Aloenxertos , Animais , Anti-Inflamatórios/administração & dosagem , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Aorta/transplante , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Citoproteção , Dopamina/administração & dosagem , Dopamina/farmacologia , Humanos , Mediadores da Inflamação/metabolismo , Bombas de Infusão Implantáveis , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neointima , Ratos Endogâmicos BN , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Tempo
17.
Transpl Int ; 29(9): 974-84, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26563531

RESUMO

Kidney transplantation is a major medical improvement for patients with end-stage renal disease, but organ shortage limits its widespread use. As a consequence, the proportion of grafts procured from extended criteria donors (ECD) has increased considerably, but this comes along with increased rates of delayed graft function (DGF) and a higher incidence of immune-mediated rejection that limits organ and patient survival. Furthermore, most grafts are derived from brain dead organ donors, but the unphysiological state of brain death is associated with significant metabolic, hemodynamic, and pro-inflammatory changes, which further compromise patient and graft survival. Thus, donor interventions to preserve graft quality are fundamental to improve long-term transplantation outcome, but interventions must not harm other potentially transplantable grafts. Several donor pretreatment strategies have provided encouraging results in animal models, but evidence from human studies is sparse, as most clinical evidence is derived from single-center or nonrandomized trials. Furthermore, ethical matters have to be considered especially concerning consent from donors, donor families, and transplant recipients to research in the field of donor treatment. This review provides an overview of clinically proven and promising preclinical strategies of donor treatment to optimize long-term results after kidney transplantation.


Assuntos
Transplante de Rim , Preservação de Órgãos/métodos , Doadores de Tecidos , Obtenção de Tecidos e Órgãos/ética , Acetilcisteína/química , Animais , Antioxidantes/metabolismo , Morte Encefálica , Desamino Arginina Vasopressina/administração & dosagem , Função Retardada do Enxerto , Dopamina/administração & dosagem , Sobrevivência de Enxerto , Humanos , Sistema Imunitário , Insulina/sangue , Falência Renal Crônica , Transplante de Rim/ética , Metilprednisolona/administração & dosagem , Ensaios Clínicos Controlados Aleatórios como Assunto , Proteínas Recombinantes/metabolismo , Ressuscitação , Superóxido Dismutase/metabolismo , Hormônios Tireóideos/sangue
18.
PLoS One ; 9(6): e99298, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24926788

RESUMO

AIM: N-acyl dopamines (NADD) are gaining attention in the field of inflammatory and neurological disorders. Due to their hydrophobicity, NADD may have access to the endoplasmic reticulum (ER). We therefore investigated if NADD induce the unfolded protein response (UPR) and if this in turn influences cell behaviour. METHODS: Genome wide gene expression profiling, confirmatory qPCR and reporter assays were employed on human umbilical vein endothelial cells (HUVEC) to validate induction of UPR target genes and UPR sensor activation by N-octanoyl dopamine (NOD). Intracellular ATP, apoptosis and induction of thermotolerance were used as functional parameters to assess adaptation of HUVEC. RESULTS: NOD, but not dopamine dose dependently induces the UPR. This was also found for other synthetic NADD. Induction of the UPR was dependent on the redox activity of NADD and was not caused by selective activation of a particular UPR sensor. UPR induction did not result in cell apoptosis, yet NOD strongly impaired cell proliferation by attenuation of cells in the S-G2/M phase. Long-term treatment of HUVEC with low NOD concentration showed decreased intracellular ATP concentration paralleled with activation of AMPK. These cells were significantly more resistant to cold inflicted injury. CONCLUSIONS: We provide for the first time evidence that NADD induce the UPR in vitro. It remains to be assessed if UPR induction is causally associated with hypometabolism and thermotolerance. Further pharmacokinetic studies are warranted to address if the NADD concentrations used in vitro can be obtained in vivo and if this in turn shows therapeutic efficacy.


Assuntos
Trifosfato de Adenosina/metabolismo , Dopamina/análogos & derivados , Células Endoteliais/efeitos dos fármacos , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Apoptose , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dopamina/farmacologia , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Técnicas In Vitro
19.
J Leukoc Biol ; 96(3): 453-62, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24929005

RESUMO

Recently, we developed a nonhemodynamic dopamine derivative, NOD, which has profound anti-inflammatory effects in vitro. As NOD also protects rats from ischemic AKI, the present study tested whether NOD is able to modulate cellular immunity for potential use as a T cell-suppressive agent. To this end, T cells were stimulated by anti-CD3/CD28 or PMA/ionomycin in the presence or absence of different concentrations of NOD. T cell proliferation, activation markers, intracellular cytokine expression, and activation of transcription factors were assessed. Whereas T cell proliferation was inhibited significantly by NOD at Day 3, proliferation was restored at Day 7 or later depending on the NOD concentration used. Inhibition of proliferation was reflected by a diminished CD25 expression and switch from naive to memory T cells. Early TCR activation events were unaffected, yet NF-κB and AP-1 were strongly inhibited by NOD. The inhibitory effect of NOD seemed to be dependent on its redox activity, as NOT, a redox-inactive NOD derivate, did not influence proliferation. NOD displayed synergistic effects with CNIs on T cell proliferation. Our data demonstrate that NOD displays T cell-suppressive activity. In keeping with its anti-inflammatory action and its beneficial effect on ischemia-induced AKI, NOD may be an interesting drug candidate to prevent CNI-related side-effects.


Assuntos
Dopamina/análogos & derivados , Fase G1/efeitos dos fármacos , Imunossupressores/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Fatores de Transcrição/antagonistas & inibidores , Inibidores de Calcineurina/farmacologia , Divisão Celular/efeitos dos fármacos , Dopamina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Humanos , Memória Imunológica , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Oxirredução , Estresse Oxidativo , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Complexo Receptor-CD3 de Antígeno de Linfócitos T/efeitos dos fármacos , Complexo Receptor-CD3 de Antígeno de Linfócitos T/imunologia , Linfócitos T/citologia , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo , Proteína-Tirosina Quinase ZAP-70/metabolismo
20.
Free Radic Biol Med ; 52(4): 794-802, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22210380

RESUMO

Carbon monoxide (CO) abrogates TNF-α-mediated inflammatory responses in endothelial cells, yet the underlying mechanism thereof is still elusive. We have previously shown that the anti-inflammatory effect of CO-releasing molecule-3 (CORM-3) is not completely mediated via deactivation of the NF-κB pathway. In this study, we sought to explore other potential mechanisms by which CORM-3 downregulates VCAM-1 expression on TNF-α-stimulated HUVECs. By genome-wide gene expression profiling and pathway analysis we studied the relevance of particular pathways for the anti-inflammatory effect of CORM-3. In CORM-3-stimulated HUVECs significant changes in expression were found for genes implicated in the proteasome and porphyrin pathways. Although proteasome activities were increased by CORM-3, proteasome inhibitors did not abolish the effect of CORM-3. Likewise, heme oxygenase-1 inhibitors did not abrogate the ability of CORM-3 to downregulate VCAM-1 expression. Interestingly, CORM-3 inhibited MAPK p38, and the p38 inhibitor SB203580 downregulated VCAM-1 expression. However, downregulation of VCAM-1 by CORM-3 occurred only at concentrations that partly inhibit ATP production and sodium azide and oligomycin paralleled the effect of CORM-3 in this regard. Our results indicate that CORM-3-induced downregulation of VCAM-1 is mediated via p38 inhibition and mitochondrial respiration, whereas the ubiquitin-proteasome system seems not to be involved.


Assuntos
Anti-Inflamatórios/farmacologia , Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases , Mitocôndrias/metabolismo , Compostos Organometálicos/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Monóxido de Carbono/farmacologia , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Regulação para Baixo , Ativação Enzimática , Perfilação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitocôndrias/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Consumo de Oxigênio , Complexo de Endopeptidases do Proteassoma/genética , Proteólise , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Ubiquitinadas/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA