Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Front Neurol ; 13: 952699, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36330424

RESUMO

Background and aim: Inflammatory myopathies are heterogeneous in terms of etiology, (immuno)pathology, and clinical findings. Endothelial cell injury, as it occurs in DM, is a common feature of numerous inflammatory and non-inflammatory vascular diseases. Vascular regeneration is mediated by both local and blood-derived mechanisms, such as the mobilization and activation of so-called proangiogenic cells (PACs) or early endothelial progenitor cells (eEPCs). The current study aimed to evaluate parameters of eEPC integrity in dermatomyositis (DM), compared to necrotizing myopathy (NM) and to non-myopathic controls. Methods: Blood samples from DM and NM patients were compared to non-myositis controls and analyzed for the following parameters: circulating CD133+/VEGFR-2+ cells, number of colony-forming unit endothelial cells (CFU-ECs), concentrations of angiopoietin 1, vascular endothelial growth factor (VEGF), and CXCL-16. Muscle biopsies from DM and NM subjects underwent immunofluorescence analysis for CXCR6, nestin, and CD31 (PECAM-1). Finally, myotubes, derived from healthy donors, were stimulated with serum samples from DM and NM patients, subsequently followed by RT-PCR for the following candidates: IL-1ß, IL-6, nestin, and CD31. Results: Seventeen (17) DM patients, 7 NM patients, and 40 non-myositis controls were included. CD133+/VEGFR-2+ cells did not differ between the groups. Both DM and NM patients showed lower CFU-ECs than controls. In DM, intramuscular CD31 abundances were significantly reduced, which indicated vascular rarefaction. Muscular CXCR6 was elevated in both diseases. Circulating CXCL-16 was higher in DM and NM in contrast, compared to controls. Serum from patients with DM but not NM induced a profound upregulation of mRNS expression of CD31 and IL-6 in cultured myotubes. Conclusion: Our study demonstrates the loss of intramuscular microvessels in DM, accompanied by endothelial activation in DM and NM. Vascular regeneration was impaired in DM and NM. The findings suggest a role for inflammation-associated vascular damage in the pathogenesis of DM.

2.
Int J Nephrol ; 2022: 9432509, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35782192

RESUMO

Methods: The study was performed in a retrospective and observational manner. All adult (age 18 years or older) in-hospital subjects treated from January until December 2019 were included. CKD was diagnosed according to the KDIGO 2012 CKD Guideline. The following variables were assessed: CKD stage, quantification/analysis (yes/no) of blood pressure, proteinuria, serum phosphate, serum 25-OH-D3, ferritin and transferrin saturation, and blood gas analysis. In addition, recommendations of the following medicines were analyzed (given/not given): ACE inhibitor or sartan, phosphate binder, vitamin D3 (activated or native), iron, erythropoietin, and bicarbonate. It was also evaluated whether discharge letters contained CKD-related diagnoses or not. Results: In total, 581 individuals were included in the study. The majority of aspects related to the monitoring and therapeutic management of CKD were either considered in only a small proportion of affected individuals (e.g., quantification of PTH - 5.5%/25-OH-D3 - 6%/transferrin saturation - 13.6%) or avoided nearly at all (e.g., recommendation of erythropoietin-1%, documentation of CKD-MBD diagnosis-0.3%). A reasonable quality of care was identified concerning the blood pressure monitoring (performed in 100%) and blood gas analysis (55% of the patients received analysis). Serum phosphate was measured in 12.9%, particularly in subjects at higher CKD stages. Conclusions: The current investigation revealed poor quality of care in CKD patients treated at the Brandenburg University Hospital over the period of one year. Quality improvement must be achieved, most likely via a standardized educational program for physicians and a directer access to CKD management guidelines.

3.
Int J Nephrol ; 2021: 5549316, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33986959

RESUMO

BACKGROUND: Acute kidney injury substantially worsens the prognosis of hospitalized patients. The Brandenburg Medical School was founded in 2014, and a nephrology section was opened in summer 2017. The aim of the study was to analyze AKI epidemiology and outcomes in one of two university hospitals belonging to the medical school. The period of interest dated from January to December 2015. METHODS: The investigation was designed as a single-center, retrospective cohort study at the Brandenburg Hospital of the Brandenburg Medical School. All in-hospital patients treated between January and the end of December 2015 were included. AKI was defined as specified in the 2012 published KDIGO criteria (criteria 1 and 2). Four parameters were evaluated in particular: AKI incidence, in-hospital mortality, frequency of renal replacement therapy, and renal recovery during the stay at the hospital. RESULTS: A total number of 5,300 patients were included in the analysis. AKI was diagnosed in 490 subjects (10.1%). The in-hospital mortality was 26%. The following conditions/parameters significantly differed between survivors (s) and nonsurviving (ns) subjects: duration of in-hospital treatment (s > ns), AKI onset (outpatient vs. in-hospital) (outpatient in s > ns), dialysis due to AKI (s < ns), vasopressor administration (s < ns), and invasive ventilation (s < ns). 5.6% received dialysis therapy, and renal recovery occurred in 31% of all surviving AKI subjects. CONCLUSION: Both, the AKI incidence and the frequency of dialysis were lower than reported in the literature. However, fewer subjects recovered from AKI. These discrepant findings possibly result from the lack of prehospitalization creatinine values, the lack of follow-up data, and a generally lower awareness for the need to perform renal replacement therapy in AKI.

4.
Int J Nephrol ; 2021: 8864183, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777453

RESUMO

INTRODUCTION: Acute kidney injury (AKI) significantly worsens the prognosis of hospitalized patients. In recent years, cell-based strategies have been established as a reliable option for improving AKI outcomes in experimental AKI. Our previous studies focused on the so-called proangiogenic cells (PACs). Mechanisms that contribute to PAC-mediated AKI protection include production/secretion of extracellular vesicles (MV, microvesicles). In addition, the cells most likely act by paracrinic processes (secretome). The current study evaluated whether AKI may be preventable by the administration of either PAC-derived MV and/or the secretome alone. METHODS: AKI was induced in male C57/Bl6N mice (8-12 weeks) by bilateral renal ischemia (IRI-40 minutes). Syngeneic murine PACs were stimulated with either melatonin, angiopoietin-1 or -2, or with bone morphogenetic protein-5 (BMP-5) for one hour, respectively. PAC-derived MV and the vesicle-depleted supernatant were subsequently collected and i.v.-injected after ischemia. Mice were analyzed 48 hours later. RESULTS: IRI induced significant kidney excretory dysfunction as reflected by higher serum cystatin C levels. The only measure that improved AKI was the injection of MV, collected from native PACs. The following conditions worsened after ischemic renal function even further: MV + Ang-1, MV + BMP-5, MV + melatonin, and MV + secretome + Ang-1. CONCLUSION: Together, our data show that PAC-mediated AKI protection substantially depends on the availability of cell-derived MV. However, since previous data showed improved AKI-protection by PACs after cell preconditioning with certain mediators (Ang-1 and -2, melatonin, BMP-5), mechanisms other than exclusively vesicle-dependent mechanisms must be involved in PAC-mediated AKI protection.

5.
Odontology ; 108(3): 441-449, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32300908

RESUMO

To evaluate serum levels of the following cytokines in rheumatoid arthritis subjects with periodontal disease: Interleukin-6, -10, -17, and -23. Patients with rheumatoid arthritis frequently suffer from periodontal disease. Both diseases partly result from a dysregulated immune response. The current study aimed to quantify Interleukin-6, -10, -17, and -23 levels in rheumatoid arthritis. It should be investigated if the periodontal disease would have additional modifying effects. A total of 157 patients were included. Serum levels of IL-6, -10, -17, and -23 were measured by ELISA. Serum IL-10 increased with longer duration of morning stiffness and with higher rheumatoid factor and anti-cyclic citrullinated peptide titres. IL-10 was also elevated with longer duration of prednisolone (< 5 mg daily) and leflunomide therapy. Subjects with lower erythrocyte sedimentation rate/longer leflunomide therapy displayed more missing teeth/more clinical attachment loss. IL-17 was higher in subjects with fewer missing teeth if the following criteria were fulfilled: shorter prednisolone (< 5 mg) and methotrexate therapy, more swollen joints, longer morning stiffness. IL-23 finally was increased in subjects with higher rheumatoid factor and in those with higher periodontal probing depth/clinical attachment loss in the following situations: lower rheumatoid factor and shorter leflunomide therapy. Subjects suffering from dental/periodontal burden show an aberrant systemic cytokine availability of serum IL-6, IL-10, IL-17 and IL-23 related to disease activity and medication. This examination underlines the complexity of potential interactions between disease activity and medication related to periodontal burden.


Assuntos
Artrite Reumatoide , Doenças Periodontais , Citocinas , Humanos , Interleucina-6 , Perda da Inserção Periodontal
6.
Eur J Med Res ; 23(1): 56, 2018 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-30413175

RESUMO

BACKGROUND: Both psoriasis (Ps) and psoriasis arthritis (PsA) have been associated with increased cardiovascular risk. Also, both are characterized by increased neovascularization. Endothelial progenitor cells (EPCs) have been implicated in promoting vascular repair in ischemic diseases. The aim of the study was to correlate the EPC system with CV risk factors and with parameters of vascular stiffness in Ps and PsA. METHODS: Twenty-six healthy subjects, 30 patients with Ps, and 31 patients PsA were included in the study. eEPC regeneration was evaluated by a colony-forming assay, circulating eEPCs were measured by cytometric analysis. For vascular analysis, all subjects underwent quantification of pulse wave velocity (PWV) and augmentation index (AIX). RESULTS: Patients were categorized upon the duration of disease, severity of skin involvement (PASI-Ps), individual pain as reflected by the VAS (PsA), CRP values, and history of treatment with one or more biologicals. Regarding the eEPC system, no significant differences were observed between the respective categories. Correlation analyses between parameters of vascular stiffness (PWV and AIX) and patterns of colony formation/circulating eEPCs did not show any correlation at all. CONCLUSION: Parameters of vascular stiffness are not significantly deteriorated in Ps/PsA. Thus, pulse wave analysis may not be suitable for CVR assessment in certain autoimmune-mediated diseases. Regenerative activity of the eEPC system/circulating eEPC numbers are not altered in Ps/PsA. One may conclude that malfunctions of the eEPC are not substantially involved in perpetuating the micro-/macrovascular alterations in Ps/PsA.


Assuntos
Artrite Psoriásica/fisiopatologia , Células Progenitoras Endoteliais/fisiologia , Psoríase/fisiopatologia , Rigidez Vascular/fisiologia , Doenças Cardiovasculares/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Onda de Pulso , Medição de Risco , Fatores de Risco
7.
J Periodontal Res ; 52(4): 745-754, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28321852

RESUMO

BACKGROUND AND OBJECTIVES: The aim of this clinical cross-sectional study was to determine the level of active matrix metalloproteinase-8 (aMMP-8) and periodontal pathogenic bacteria in gingival crevicular fluid in patients with rheumatoid arthritis (RA) with varying periodontal conditions. MATERIAL AND METHODS: In total, 103 patients with RA and 104 healthy controls (HC) were included. The assessment of periodontal status included periodontal probing depth, bleeding on probing and clinical attachment loss. Periodontal disease was classified as healthy/mild, moderate or severe. For the determination of aMMP-8 levels using enzyme-linked immunosorbent assay and periodontal pathogenic bacteria using polymerase chain reaction, samples of gingival crevicular fluid were taken from the deepest gingival pockets. The statistical analyses used included a Mann-Whitney U-test, a chi-squared test or a Fisher's exact test, and the significance level was set at α = 5%. RESULTS: We found that 65% of patients with RA and 79% of HC had moderate to severe periodontal disease (p = 0.02). The prevalence of periodontal pathogens was almost equal (p > 0.05). Furthermore, depending on periodontal disease severity only minor differences in bacterial prevalence were detected. With increasing severity of periodontal disease, higher aMMP-8 levels were observed. Accordingly, a significant difference in patients with moderate periodontal disease (RA: 15.3 ± 13.8; HC: 9.1 ± 9.1; p ≤ 0.01) and severe periodontal disease (RA: 21.7 ± 13.3; HC: 13.1 ± 8.6; p = 0.07) was detected, with a greater tendency in the latter group. CONCLUSION: The increased aMMP-8 levels in the RA group indicate that the presence of RA appears to have an influence on the host response at a comparable level of bacterial load and periodontal disease severity.


Assuntos
Artrite Reumatoide/complicações , Líquido do Sulco Gengival/enzimologia , Líquido do Sulco Gengival/microbiologia , Metaloproteinase 8 da Matriz/metabolismo , Periodontite/enzimologia , Periodontite/microbiologia , Adolescente , Adulto , Idoso , Estudos Transversais , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Perda da Inserção Periodontal/microbiologia , Índice Periodontal , Bolsa Periodontal/microbiologia , Reação em Cadeia da Polimerase
8.
BMC Nephrol ; 18(1): 53, 2017 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-28166726

RESUMO

BACKGROUND: In recent years, early Endothelial Progenitor Cells (eEPCs) have been proven as effective tool in murine ischemic AKI and in diabetic nephropathy. The mechanisms of eEPC-mediated vasoprotection have been elucidated in detail. Besides producing a diverse range of humoral factors, the cells also act by secreting vasomodulatory microvesicles. Only few data in contrast have been published about the role of so-called Endothelial Colony Forming Cells (ECFCs - late EPCs) in ischemic AKI. We thus aimed to investigate ECFC effects on postischemic kidney function over several weeks. Our special interest focused on endothelial-to-mesenchymal transition (EndoMT), peritubular capillary density (PTCD), endothelial alpha-Tubulin (aT - cytoskeletal integrity), and endothelial p62 (marker of autophagocytic flux). METHODS: Eight to twelve weeks old male C57Bl/6 N mice were subjected to bilateral renal pedicle clamping for 35 or 45 min, respectively. Donor-derived syngeneic ECFCs (0.5 × 106) were i.v. injected at the end of ischemia. Animals were analyzed 1, 4 and 6 weeks later. RESULTS: Cell therapy improved kidney function exclusively at week 1 (35 and 45 min). Ischemia-induced fibrosis was diminished in all experimental groups by ECFCs, while PTCD loss remained unaffected. Significant EndoMT was detected in only two of 6 groups (35 min, week 4 and 45 min, week 6), ECFCs reduced EndoMT only in the latter. Endothelial aT declined under almost all experimental conditions and these effects were further aggravated by ECFCs. p62 was elevated in endothelial cells, more so after 45 than after 35 min of ischemia. Cell therapy did not modulate p62 abundances at any time point. CONCLUSION: A single dose of ECFCs administered shortly post-ischemia is capable to reduce interstitial fibrosis in the mid- to long-term whereas excretory dysfunction is improved only in a transient manner. There are certain differences in renal outcome parameters between eEPCs and ECFC. The latter do not prevent animals from peritubular capillary loss and they also do not further elevate endothelial p62. We conclude that differences between eEPCs and ECFCs result from certain mechanisms by which the cells act around and within vessels. Overall, ECFC treatment was not as efficient as eEPC therapy in preventing mice from ischemia-induced mid- to long-term damage.


Assuntos
Injúria Renal Aguda/terapia , Células Endoteliais/transplante , Células Progenitoras Endoteliais/transplante , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Creatinina/metabolismo , Modelos Animais de Doenças , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão/complicações , Células-Tronco , Tubulina (Proteína)/metabolismo
9.
BMC Musculoskelet Disord ; 17: 339, 2016 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-27519706

RESUMO

BACKGROUND: Patients with systemic sclerosis (SSc) are endagered by tissue fibrosis and by microvasculopathy, with the latter caused by endothelial cell expansion/proliferation. SSc-associated fibrosis potentially results from mesenchymal transdifferentiation of endothelial cells. Early Endothelial Progenitor Cells (eEPCs) act proangiogenic under diverse conditions. Aim of the study was to analyze eEPC regeneration and mesenchymal transdifferentiation in patients with limited and diffuse SSs (lSSc and dSSc). METHODS: Patients with both, lSSc and dSSc were included into the study. The following parameters were evaluated: eEPC numbers and regeneration, concentrations of vasomodulatory mediators, mesenchymal properties of blood-derived eEPC. Serum samples of healthy subjects and SS patients were used for stimulation of cultured human eEPC, subsequently followed by analysis of mesenchymal cell characteristics and mobility. RESULTS: Twenty-nine patients were included into the study. Regenerative activity of blood-derived eEPCs did not differ between Controls and patients. Circulating eEPC were significantly lower in all patients with SSc, and in limited and diffuse SSc (lSSc/dSSc). Serum concentrations of promesenchymal TGF-b was elevated in all patients with SSc. Cultured mononuclear cells from SS patients displayed higher abundances of CD31 and of CD31 and aSMA combined. Finally, serum from SSc patients inhibited migration of cultured eEPCs and the cells showed lower sensitivity towards the endothelin antagonist Bosentan. CONCLUSIONS: The eEPC system, which represents an essential element of the endogenous vascular repair machinery is affected in SSc. The increased appearance of mesenchymal properties in eEPC may indicate that alterations of the cells potentially contribute to the accumulation of connective tissue and to vascular malfunction.


Assuntos
Transdiferenciação Celular , Células Progenitoras Endoteliais/fisiologia , Esclerodermia Difusa/etiologia , Esclerodermia Limitada/etiologia , Biomarcadores/sangue , Estudos de Casos e Controles , Movimento Celular , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Regeneração , Esclerodermia Difusa/sangue , Esclerodermia Limitada/sangue
10.
Am J Physiol Renal Physiol ; 310(7): F679-F687, 2016 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-26792062

RESUMO

Renal ischemia induces peritubular capillary rarefication and fibrosis, with the latter partly resulting from the endothelial-to-mesenchymal transition (EndoMT). Endothelial cilia transmit blood flow-associated forces into the cell. Early endothelial progenitor cells (eEPCs) have been shown to protect mice from acute kidney injury in the short term. The aim of the present study was to analyze midterm consequences of eEPC treatment in the context of endothelial cilia and the EndoMT. Male C57/Bl6N mice were subjected to unilateral renal ischemia postuninephrectomy. Syngeneic murine eEPCs were systemically injected at the time of reperfusion. Animals were investigated 1, 4, and 6 wk later. Cultured mature endothelial cells were exposed to a variable flow with versus without eEPC supernatant incubation. Systemically injected eEPCs reduced serum creatinine levels at week 1 (35 and 45 min) and week 4 (45 min). Interstitial fibrosis was significantly diminished by cell treatment at all time points as well. The EndoMT was less pronounced at week 4 (35 min) and week 6 (45 min). eEPC supernatant reduced α-smooth muscle actin expression and α-tubulin abundance in flow-treated cultured mature endothelial cells, and percentages of cilium-positive cells increased. The loss of peritubular capillaries was prevented by eEPCs. Intrarenal endothelial α-tubulin decreased postischemia and was further reduced by eEPC administration. We conclude that eEPCs are capable of reorganizing the endothelial cytoskeleton in an indirect manner, ultimately resulting in stabilization of the endothelial ciliome. The investigation indicates an antimesenchymal role of endothelial cilia in the process of postischemic tissue fibrosis/EndoMT.


Assuntos
Injúria Renal Aguda/metabolismo , Cílios/metabolismo , Células Endoteliais/metabolismo , Isquemia/metabolismo , Rim/irrigação sanguínea , Traumatismo por Reperfusão/metabolismo , Células-Tronco/metabolismo , Animais , Masculino , Camundongos
12.
Am J Physiol Renal Physiol ; 307(6): F686-94, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25080521

RESUMO

Diabetic nephropathy is the most frequent single cause of end-stage renal disease in our society. Microvascular damage is a key event in diabetes-associated organ malfunction. Early endothelial outgrowth cells (eEOCs) act protective in murine acute kidney injury. The aim of the present study was to analyze consequences of eEOC treatment of murine diabetic nephropathy with special attention on endothelial-to-mesenchymal transdifferentiation, autophagy, senescence, and apoptosis. Male C57/Bl6N mice (8-12 wk old) were treated with streptozotocin for 5 consecutive days. Animals were injected with untreated or bone morphogenetic protein (BMP)-5-pretreated syngeneic murine eEOCs on days 2 and 5 after the last streptozotocin administration. Four, eight, and twelve weeks later, animals were analyzed for renal function, proteinuria, interstitial fibrosis, endothelial-to-mesenchymal transition, endothelial autophagy, and senescence. In addition, cultured mature murine endothelial cells were investigated for autophagy, senescence, and apoptosis in the presence of glycated collagen. Diabetes-associated renal dysfunction (4 and 8 wk) and proteinuria (8 wk) were partly preserved by systemic cell treatment. At 8 wk, antiproteinuric effects were even more pronounced after the injection of BMP-5-pretreated cells. The latter also decreased mesenchymal transdifferentiation of the endothelium. At 8 wk, intrarenal endothelial autophagy (BMP-5-treated cells) and senescence (native and BMP-5-treated cells) were reduced. Autophagy and senescence in/of cultured mature endothelial cells were dramatically reduced by eEOC supernatant (native and BMP-5). Endothelial apoptosis decreased after incubation with eEOC medium (native and BMP-5). eEOCs act protective in diabetic nephropathy, and such effects are significantly stimulated by BMP-5. The cells modulate endothelial senescence, autophagy, and apoptosis in a protective manner. Thus, the renal endothelium could serve as a therapeutic target in diabetes-associated kidney dysfunction.


Assuntos
Proteína Morfogenética Óssea 5/fisiologia , Nefropatias Diabéticas/terapia , Células Endoteliais/transplante , Animais , Apoptose , Autofagia , Transdiferenciação Celular , Senescência Celular , Células Endoteliais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteinúria/prevenção & controle
13.
Lupus ; 22(14): 1523-5, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24014569

RESUMO

We report the case of a 19-year-old woman with progressive proliferative lupus nephritis (LN) class III after induction and maintenance therapy with mycophenolate mofetil (MMF). Despite a satisfying clinical improvement proteinuria progressed under this medication. We treated the patient with additional belimumab after discussing other options. Following treatment with belimumab, proteinuria rapidly improved to almost normal levels and clinical remission lasted. Belimumab might hold promise for this indication.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Imunossupressores/uso terapêutico , Nefrite Lúpica/tratamento farmacológico , Progressão da Doença , Feminino , Humanos , Nefrite Lúpica/fisiopatologia , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapêutico , Proteinúria/tratamento farmacológico , Proteinúria/etiologia , Indução de Remissão , Resultado do Tratamento , Adulto Jovem
14.
J Transplant ; 2012: 764154, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22530109

RESUMO

Acute renal failure (ARF) severely worsens prognosis of hospitalized patients. The most frequent cause of intrarenal ARF is transient or prolonged renal hypoperfusion (ischemia). Ischemia primarily affects the function and structure of tubular epithelial cells, which, in severe cases, is characterized by epithelial cell necrosis. Nevertheless, ischemia does not exclusively lead to alterations of epithelial cells but also causes interstitial inflammation and interstitial microvasculopathy. Both inflammation and microvasculopathy are particularly important in terms of postischemic kidney repair. Postischemic microvasculopathy is characterized by endothelial cell swelling with subsequent microvascular occlusion. Thus, reperfusion is inhibited (no-reflow phenomenon). Such endothelial cell dysfunction offers new therapeutic perspectives in ischemic ARF. Newer observations point towards the role of the so-called endothelial progenitor cells (EPCs) in the treatment of ARF. Systemic administration of EPCs to mice with bilateral renal ischemia mitigates postischemic endothelial cell dysfunction and protects animals from acute renal failure.

15.
Am J Physiol Renal Physiol ; 302(10): F1305-12, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22357919

RESUMO

Endothelial progenitor cells (EPCs) protect the kidney from acute ischemic injury. The aim of this study was to analyze whether pretreatment of murine "early outgrowth" EPCs (eEPCs) with the hormone melatonin increases the cells' renoprotective effects in the setting of murine acute ischemic renal failure. Male (8-12 wk old) C57Bl/6N mice were subjected to unilateral ischemia-reperfusion injury postuninephrectomy (40 min). Postischemic animals were injected with either 0.5×10(6) untreated syngeneic murine eEPCs or with cells, pretreated with melatonin for 1 h. Injections were performed shortly after reperfusion of the kidney. While animals injected with untreated cells developed acute renal failure, eEPC pretreatment with melatonin dramatically improved renoprotective actions of the cells. These effects were completely reversed after cell pretreatment with melatonin and the MT-1/-2 antagonist luzindole. In vitro analysis revealed that melatonin reduced the amount of tumor growth factor-ß-induced eEPC apoptosis/necrosis. Secretion of vascular endothelial growth factor by the cells was markedly stimulated by the hormone. In addition, migratory activity of eEPCs was enhanced by melatonin and supernatant from melatonin-treated eEPCs stimulated migration of cultured mature endothelial cells. In summary, melatonin was identified as a new agonist of eEPCs in acute ischemic kidney injury.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Células Endoteliais/citologia , Melatonina/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Células-Tronco/efeitos dos fármacos , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Necrose , Neovascularização Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Fator de Crescimento Transformador beta/metabolismo
16.
Panminerva Med ; 54(1): 45-52, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22278116

RESUMO

Ischemic acute kidney injury (iAKI) is the most frequent type of hospital-aquired acute renal failure (ARF). Mortality of ARF still ranges between 30% and 50%. Although acute renal ischemia significantly affects function and structure of the tubular epithelium, postischemic interstitial inflammation and microvasculopathy both contribute to ongoing renal dysfunction. The renal vasculature shows a highly specialized architecture. Renal ischemia induces severe alterations of the endothelium in small peritubular arterioles and capillaries. Investigations performed in recent years point towards a new therapeutic approach in iAKI, that is to target postischemic endothelial dysfunction by administering cells of the endothelial lineage. With regard to endothelial-type cells, new perspectives are offered with the identificaton of endothelial progenitor cells (EPCs). Although heterogenous in nature, EPCs can be employed for anti-ischemic treatment in different situations. Meanwhile the cells have been shown to protect mice from iAKI and several strategies have been established in order to increase the renoprotective capacity of EPCs. Further investigations will help to clarify whether EPCs not only protect the kidney in the short- but also in the long-term.


Assuntos
Injúria Renal Aguda/cirurgia , Células Endoteliais/transplante , Isquemia/cirurgia , Rim/irrigação sanguínea , Microvasos/patologia , Transplante de Células-Tronco , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Células Endoteliais/patologia , Humanos , Isquemia/complicações , Isquemia/patologia , Isquemia/fisiopatologia , Rim/patologia , Rim/fisiopatologia , Microcirculação , Microvasos/fisiopatologia , Circulação Renal
17.
Int J Nephrol ; 2011: 828369, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21603112

RESUMO

Acute ischemic kidney injury is the most frequent cause of acute renal failure in daily clinical practice. It has become increasingly recognized that microvascular endothelial cell dysfunction (ED) in peritubular capillaries inhibits the process of postischemic renal reperfusion. ED can serve as therapeutic target in the management of acute ischemic kidney injury. Postischemic reflow can be restored by systemic administration of either mature endothelial cells or of endothelial progenitor cells. Endothelial progenitor cells EPCs can be cultured from the peripheral circulation of humans and different animals. The cells act vasoprotectively by direct and indirect mechanisms. The protective effects of EPCs in acute ischemic kidney injury can be stimulated by preincubating the cells with different agonistic mediators. This paper summarizes the currently available data on strategies to improve the renoprotective activity of EPCs in acute ischemic kidney injury.

18.
Am J Physiol Renal Physiol ; 298(1): F78-85, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19906949

RESUMO

Endothelial progenitor cells (EPCs) protect kidneys from acute ischemic damage. The aim of this study was to identify "treatment parameters" that optimize an EPC-based therapy of acute ischemic renal failure. Male C57BL/6N mice underwent unilateral nephrectomy with simultaneous contralateral renal artery clamping for 30, 35, and 40 min. Tagged murine EPCs were systemically injected at the time of reperfusion. In some experiments, EPCs were pretreated with the Epac (exchange protein directly activated by cAMP-1) activator 8-pCPT-2'-O-Me-cAMP (Epac-1 Ac) and the integrin binding antagonist cyclic Arg-Gly-Asp peptide (cRGD). Injections of 10(6) EPCs after 30 and 35 min of renal ischemia protected animals from acute renal failure. The same effect occurred with 0.5 x 10(6) EPCs after a 35-min period of ischemia. If ischemia lasted for 40 min, 0.5 x 10(6) cells mice did not prevent acute renal failure. To analyze whether EPC integrin receptor activation would modify the cells' renoprotective activity, EPCs were pretreated with Epac-1 Ac. Such animals did not develop acute renal failure, even if ischemia lasted for 40 min. This effect was negated if the cells were pretreated with both Epac-1 Ac and cRGD. In kidneys from those animals medullopapillary EPCs were significantly accumulated. In vitro Epac-1 Ac preactivation of EPCs did not increase the overall expression intensity but induced a redistribution of beta(1)-integrins toward the cell membranes. We conclude that EPC pretreatment with the integrin receptor activator 8-pCPT-2'-O-Me-cAMP augments the anti-ischemic potential of the cells.


Assuntos
Injúria Renal Aguda/prevenção & controle , AMP Cíclico/análogos & derivados , Células Endoteliais/metabolismo , Células Endoteliais/transplante , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Células-Tronco/metabolismo , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , AMP Cíclico/uso terapêutico , Modelos Animais de Doenças , Células Endoteliais/patologia , Cadeias beta de Integrinas/metabolismo , Rim/irrigação sanguínea , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nefrectomia , Peptídeos Cíclicos/uso terapêutico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transplante de Células-Tronco/métodos , Células-Tronco/patologia , Transplante Homólogo
19.
Scand J Immunol ; 66(5): 584-90, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17868260

RESUMO

B-cell-dependent autoantibody production is a hallmark of systemic lupus erythematosus (SLE) which requires costimulatory molecules. The aim of the study was to analyse the expression of costimulatory molecules on B cells in patients with SLE. Twenty-six patients with SLE (four male, 22 female, mean age 46 +/- 15 years) as defined by the American College of Rheumatology criteria and 13 healthy controls (three male, 10 female, mean age 43 +/- 15 years) were included in the study. In a subgroup analysis, SLE patients were divided according to renal involvement due to SLE (10 with and 16 patients without renal involvement). Clinical disease activity was assessed according to the systemic lupus erythematosus disease activity index (SLEDAI). Blood B-cell populations were analysed by FACS for the cell surface marker expression of CD27, CD38, CD71, CD80, CD86 and CD137 ligand. The expression levels of CD71, CD80 and CD86 on B cells were significantly enhanced in SLE patients when compared with healthy controls (27 +/- 3% versus 11 +/- 2%, P = 0.0003, 55 +/- 2% versus 28 +/- 4%, P < 0.0001, 34 +/- 3% versus 12 +/- 2%, P < 0.0001). CD86 expression was significantly elevated in patients with renal involvement when compared with patients without renal disease (43 +/- 6% versus 28 +/- 3%, P < 0.05). There was a significant correlation between the expression levels of CD80 and CD86 on CD19(+) B cells and disease activity. Moreover, prednisone dose significantly correlated with SLEDAI (r = 0.5, P = 0.02) and with the expression levels of CD86 (r = 0.47, P = 0.02). A pathological B-cell population is associated with disease activity and renal involvement in SLE which are obviously resistant to therapy with medium doses of prednisone.


Assuntos
Antígenos CD/biossíntese , Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Nefrite Lúpica/imunologia , Ativação Linfocitária/imunologia , Adulto , Idoso , Subpopulações de Linfócitos B/metabolismo , Linfócitos B/metabolismo , Feminino , Citometria de Fluxo , Humanos , Lúpus Eritematoso Sistêmico/metabolismo , Nefrite Lúpica/metabolismo , Masculino , Pessoa de Meia-Idade , Fenótipo , Prognóstico
20.
Clin Exp Immunol ; 145(2): 235-42, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16879242

RESUMO

Systemic lupus erythematosus (SLE) is characterized by a deviation of the immune system that involves T cell-dependent autoantibody production. The aim of this study was to investigate the role of co-stimulatory markers on T cells in this disease. Twenty-eight patients with SLE as defined by the American College of Rheumatology (ACR) criteria and 11 healthy controls were included into the study. Eleven patients had biopsy-proven lupus nephritis while 17 patients had no clinical evidence of lupus nephritis. Clinical disease activity was assessed according to the systemic lupus erythematosus disease index (SLEDAI). CD4+ T cell populations in the peripheral blood were analysed for the expression of co-stimulatory markers CD45RO, CD70, CD80, CD86, CD137, CD137L, CD134, CD152, CD154 and ICOS. SLE patients showed an increased frequency of peripheral CD4+ T cells expressing high levels of CD80, CD86 and CD134 compared to healthy controls (7.1 +/- 1.5% versus 1.7 +/- 0.9%; P < 0.005; 2.3 +/- 0.4% versus 1.0 +/- 0.2%; P = 0.008, 20.2 +/- 2.0% versus 10.6 +/- 1.9%; P < 0.005, respectively). Significantly higher levels of CD80 on CD4+ T cells were detected in SLE patients with lupus nephritis compared to patients without nephritis (11.9 +/- 3.3% versus 4.0 +/- 0.7%; P < 0.005). There was an increased presence of CD134+ CD4+ cells in SLE patients with lupus nephritis (27.5 +/- 4.0% versus 15.5 +/- 1.3%; P < 0.005). CD80 and CD134 expression was significantly correlated with SLEDAI (r = 0.42, P = 0.03; r = 0.56, P < 0.005). Co-stimulatory molecules on CD4+ T cells are associated with renal disease and disease activity in patients with systemic lupus erythematosus.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Nefrite/imunologia , Receptores do Fator de Necrose Tumoral/análise , Doença Aguda , Adulto , Reações Antígeno-Anticorpo , Antígeno B7-1/análise , Biomarcadores/análise , Feminino , Citometria de Fluxo/métodos , Humanos , Imunossupressores/uso terapêutico , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Subpopulações de Linfócitos/imunologia , Masculino , Glicoproteínas de Membrana/imunologia , Microscopia Confocal/métodos , Pessoa de Meia-Idade , Nefrite/complicações , Nefrite/tratamento farmacológico , Ligante OX40 , Receptores OX40 , Receptores do Fator de Necrose Tumoral/metabolismo , Estatísticas não Paramétricas , Transtornos Relacionados ao Uso de Substâncias , Fatores de Necrose Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...