Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
World J Surg ; 48(8): 1958-1966, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38877383

RESUMO

BACKGROUND: In June 2021, the first robot-assisted donor nephrectomy (RADN) was performed at the Leiden University Medical Center (LUMC), the Netherlands. The goal of this study was to investigate whether this procedure has been implemented safely and efficiently. METHODS: RADN was retrospectively compared to laparoscopic donor nephrectomy (LDN) performed during the same time period (June 2021 until November 2022). Patients were assigned to RADN depending on the availability of the da Vinci robot and surgical team. The studied endpoints were postoperative complications, operative time, estimated blood loss, warm ischemic time (WIT), and postoperative pain experience. For analysis, the Student's t-test and Chi-squared test were used for, respectively, continuous and categorical data. RESULTS: Forty RADN were compared to 63 LDN. Total insufflation time was significantly longer in RADN compared to LDN (188 min (169-214) versus 172 min (144-194); p = 0.02). Additionally, WIT was also found to be significantly higher in the robot-assisted group (04:54 min vs. 04:07 min; p < 0.01). No statistical differences were found in postoperative outcomes (eGFR of the recipient at 3-month follow-up, RADN 54.08 mL/min ±18.79 vs. LDN 56.41 mL/min ±16.82; p = 0.52), pain experience, and complication rate. CONCLUSION: RADN was safely and efficiently implemented at the LUMC. It's results were not inferior to laparoscopic donor nephrectomy. Operative time and warm ischemic times were longer in RADN. This may relate to a learning curve effect. No clinically relevant effect on postoperative outcomes was observed.


Assuntos
Laparoscopia , Doadores Vivos , Nefrectomia , Procedimentos Cirúrgicos Robóticos , Humanos , Nefrectomia/métodos , Procedimentos Cirúrgicos Robóticos/métodos , Feminino , Masculino , Laparoscopia/métodos , Pessoa de Meia-Idade , Estudos Retrospectivos , Adulto , Transplante de Rim/métodos , Coleta de Tecidos e Órgãos/métodos , Duração da Cirurgia , Competência Clínica , Resultado do Tratamento , Países Baixos , Idoso
2.
Ann Surg ; 270(5): 877-883, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31567503

RESUMO

OBJECTIVE: To explore putative different impacts of delayed graft function (DGF) on long-term graft survival in kidneys donated after brain death (DBD) and circulatory death (DCD). BACKGROUND: Despite a 3-fold higher incidence of DGF in DCD grafts, large studies show equivalent long-term graft survival for DBD and DCD grafts. This observation implies a differential impact of DGF on DBD and DCD graft survival. The contrasting impact is remarkable and yet unexplained. METHODS: The impact of DGF on DBD and DCD graft survival was evaluated in 6635 kidney transplants performed in The Netherlands. DGF severity and functional recovery dynamics were assessed for 599 kidney transplants performed at the Leiden Transplant Center. Immunohistochemical staining, gene expression profiling, and Ingenuity Pathway Analysis were used to identify differentially activated pathways in DBD and DCD grafts. RESULTS: While DGF severely impacted 10-year graft survival in DBD grafts (HR 1.67; P < 0.001), DGF did not impact graft survival in DCD grafts (HR 1.08; P = 0.63). Shorter dialysis periods and superior posttransplant eGFRs in DBD grafts show that the differential impact was not caused by a more severe DGF phenotype in DBD grafts. Immunohistochemical evaluation indicates that pathways associated with tissue resilience are present in kidney grafts. Molecular evaluation showed selective activation of resilience-associated pathways in DCD grafts. CONCLUSIONS: This study shows an absent impact of DGF on long-term graft survival in DCD kidneys. Molecular evaluation suggests that the differential impact of DGF between DBD and DCD grafts relates to donor-type specific activation of resilience pathways in DCD grafts.


Assuntos
Função Retardada do Enxerto/fisiopatologia , Falência Renal Crônica/cirurgia , Transplante de Rim/mortalidade , Transplante de Rim/métodos , Sistema de Registros , Idoso , Análise de Variância , Morte Encefálica , Função Retardada do Enxerto/mortalidade , Estudos de Avaliação como Assunto , Feminino , Rejeição de Enxerto , Sobrevivência de Enxerto , Insuficiência Cardíaca/mortalidade , Humanos , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/mortalidade , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Países Baixos , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Medição de Risco , Fatores de Tempo , Doadores de Tecidos
3.
Am J Transl Res ; 10(3): 816-826, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29636871

RESUMO

The pathophysiology of ischemia/reperfusion (I/R) injury is complex and poorly understood. Animal studies imply platelet activation as an initiator of the inflammatory response upon reperfusion. However, it remains unclear whether and how these results translate to clinical I/R. This study evaluates putative platelet activation in the context of two forms of clinical I/R (heart valve surgery with aortic-cross clamping, n = 39 and kidney transplantation, n = 34). The technique of sequential selective arteriovenous (AV) measurements over the reperfused organs was applied to exclude the influence of systemic changes occurring during surgery while simultaneously maximizing sensitivity. Platelet activation and degranulation was evaluated by assessing the expression levels of established markers, i.e. RANTES (Regulated on Activation, Normal T Cell Expressed and Secreted), ß-thromboglobulin (ß-TG), platelet-derived growth factor (PDGF)-BB and CXCL8 (known as interleukin-8), and by employing an in-vitro assay that specifically tests for platelet excitability. Moreover, a histological analysis was performed by means of CD41 staining. Results show stable RANTES, ß-TG, PDGF-BB and CXCL8 AV-concentrations within the first half hour over the reperfused organs, suggesting that myocardial and renal I/R are not associated with platelet activation. Results from the platelet excitability assay were in line with these findings and indicated reduced and stable platelet excitability following renal and myocardial reperfusion, respectively. Histological analysis yield evidence of platelet marginalization in the reperfused organs. In conclusion, results from this study do not support a role for platelet activation in early phases of clinical I/R injury.

4.
Kidney Int ; 90(1): 181-91, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27188504

RESUMO

Delayed graft function (DGF) following kidney transplantation affects long-term graft function and survival and is considered a manifestation of ischemia reperfusion injury. Preclinical studies characterize metabolic defects resulting from mitochondrial damage as primary driver of ischemia reperfusion injury. In a comprehensive approach that included sequential establishment of postreperfusion arteriovenous concentration differences over the human graft, metabolomic and genomic analysis in tissue biopsies taken before and after reperfusion, we tested whether the preclinical observations translate to the context of clinical DGF. This report is based on sequential studies of 66 eligible patients of which 22 experienced DGF. Grafts with no DGF immediately recovered aerobic respiration as indicated by prompt cessation of lactate release following reperfusion. In contrast, grafts with DGF failed to recover aerobic respiration and showed persistent adenosine triphosphate catabolism indicated by a significant persistently low post reperfusion tissue glucose-lactate ratio and continued significant post-reperfusion lactate and hypoxanthine release (net arteriovenous difference for lactate and hypoxanthine at 30 minutes). The metabolic data for the group with DGF point to a persistent post reperfusion mitochondrial defect, confirmed by functional (respirometry) and morphological analyses. The archetypical mitochondrial stabilizing peptide SS-31 significantly preserved mitochondrial function in human kidney biopsies following simulated ischemia reperfusion. Thus, development of DGF is preceded by a profound post-reperfusion metabolic deficit resulting from severe mitochondrial damage. Strategies aimed at preventing DGF should be focused on safeguarding a minimally required post-reperfusion metabolic competence.


Assuntos
Aloenxertos/patologia , Função Retardada do Enxerto/metabolismo , Sobrevivência de Enxerto , Transplante de Rim/efeitos adversos , Mitocôndrias/patologia , Traumatismo por Reperfusão/complicações , Aloenxertos/metabolismo , Biópsia , Estudos de Coortes , Função Retardada do Enxerto/epidemiologia , Função Retardada do Enxerto/etiologia , Função Retardada do Enxerto/patologia , Feminino , Humanos , Incidência , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Masculino , Pessoa de Meia-Idade , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Oligopeptídeos/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
5.
Microrna ; 3(3): 144-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25541911

RESUMO

Integrity of the capillary network in the kidney is essential in the recovery from ischemia/ reperfusion injury (IRI), a phenomenon central to kidney transplantation and acute kidney injury. MicroRNA- 126 (miR-126) is known to be important in maintaining vascular homeostasis by facilitating vascular regeneration and modulating the mobilization of vascular progenitor cells. Stromal cell-derived factor 1 (SDF-1), important in the mobilization of vascular progenitor cells, is a direct target of miR-126 and modulation of miR-126 was previously shown to affect the number of circulating Sca-1(+)/Lin(-) vascular progenitor cells in a mouse model for hind limb ischemia. Here, we assessed the in vivo contribution of miR-126 to progenitor cell mobilization and kidney function following IRI in mice. A three day follow up of blood urea levels following kidney IRI demonstrated that systemic antagomir silencing of miR-126 did not impact the loss or subsequent restoration of kidney function. However, whole kidney lysates displayed elevated gene expression levels of Sdf-1, Vegf-A and eNOS after IRI as a result of systemic silencing of miR-126. Furthermore, FACS-analysis on whole blood three days after surgery revealed a marked up regulation of the number of circulating Sca-1(+)/Lin(-) progenitor cells in the antagomir-126 treated mice, in an ischemia dependent manner. Our data indicate that silencing of miR-126 can enhance renal expression of Sdf-1 after IRI, leading to the mobilization of vascular progenitor cells into the circulation.


Assuntos
Antígenos Ly/metabolismo , Quimiocina CXCL12/metabolismo , Células Progenitoras Endoteliais/metabolismo , Rim/metabolismo , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Antígenos Ly/genética , Quimiocina CXCL12/genética , Células Progenitoras Endoteliais/citologia , Inativação Gênica , Rim/irrigação sanguínea , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Cytotherapy ; 15(6): 663-72, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23419679

RESUMO

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) are pluripotent cells that have immunosuppressive and reparative properties in vitro and in vivo. Although autologous bone marrow (BM)-derived MSCs are already clinically tested in transplant recipients, it is unclear whether these BM cells are affected by renal disease. We assessed whether renal failure affected the function and therapeutic potential of BM-MSCs. METHODS: MSCs from 10 adults with end-stage renal disease (ESRD) and 10 age-matched healthy controls were expanded from BM aspirates and tested for phenotype and functionality in vitro. RESULTS: MSCs from ESRD patients were >90% positive for CD73, CD90 and CD105 and negative for CD34 and CD45 and showed a similar morphology and differentiation capacity as MSCs from healthy controls. Of importance for their clinical utility, growth characteristics were similar in both groups, and sufficient numbers of MSCs were obtained within 4 weeks. Messenger RNA expression levels of self-renewal genes and factors involved in repair and inflammation were also comparable between both groups. Likewise, microRNA expression profiling showed a broad overlap between ESRD and healthy donor MSCs. ESRD MSCs displayed the same immunosuppressive capacities as healthy control MSCs, demonstrated by a similar dose-dependent inhibition of peripheral blood mononuclear cell proliferation, similar inhibition of proinflammatory cytokines tumor necrosis factor-α and interferon-γ production and a concomitant increase in the production of interleukin-10. CONCLUSIONS: Expanded BM-MSCs procured from ESRD patients and healthy controls are both phenotypically and functionally similar. These findings are important for the potential autologous clinical application of BM-MSCs in transplant recipients.


Assuntos
Células da Medula Óssea/citologia , Terapia Baseada em Transplante de Células e Tecidos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Idoso , Feminino , Humanos , Falência Renal Crônica/patologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Transplante Autólogo
7.
Transplantation ; 95(6): 816-20, 2013 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-23348894

RESUMO

BACKGROUND: Ischemia/reperfusion (I/R) injury has a major impact on kidney graft function and survival. Animal studies have suggested a role for complement activation in mediating I/R injury; however, results are not unambiguous. Whether complement activation is involved in clinical I/R injury in humans is still unclear. METHODS: In the present study, we assessed the formation and release of C5b-9 during early reperfusion in clinical kidney transplantation in living donor, brain-dead donor, and cardiac dead donor kidney transplantation. By arteriovenous measurements and histologic studies, local terminal complement activation in the reperfused kidney was assessed. RESULTS: There was no release of soluble C5b-9 (sC5b-9) from living donor kidneys, nor was there a release of C5a. In contrast, instantly after reperfusion, there was a significant but transient venous release of sC5b-9 from the reperfused kidney graft in brain-dead donor and cardiac dead donor kidney transplantation. This short-term activation of the terminal complement cascade in deceased-donor kidney transplantation was not reflected by renal tissue deposition of C5b-9 in biopsies taken 45 min after reperfusion. CONCLUSIONS: This systematic study in human kidney transplantation shows an acute but nonsustained sC5b-9 release on reperfusion in deceased-donor kidney transplantation. This instantaneous, intravascular terminal complement activation may be induced by intravascular cellular debris and hypoxic or injured endothelium.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Transplante de Rim/métodos , Reperfusão/métodos , Doença Aguda , Adulto , Idoso , Animais , Biópsia/métodos , Ativação do Complemento , Feminino , Humanos , Imuno-Histoquímica/métodos , Rim/anatomia & histologia , Masculino , Pessoa de Meia-Idade , Modelos Estatísticos , Traumatismo por Reperfusão/terapia , Doadores de Tecidos
8.
Stem Cells Transl Med ; 2(2): 107-11, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23349326

RESUMO

Despite excellent short-term results, long-term survival of transplanted kidneys has not improved accordingly. Although alloimmune responses and calcineurin inhibitor-related nephrotoxicity have been identified as main drivers of fibrosis, no effective treatment options have emerged. In this perspective, mesenchymal stromal cells (MSCs) are an interesting candidate because of their immunosuppressive and regenerative properties. Of importance, no other clinical studies have investigated their effects in allograft rejection and fibrosis. We performed a safety and feasibility study in kidney allograft recipients to whom two intravenous infusions (1 million cells per kilogram) of autologous bone marrow (BM) MSCs were given, when a protocol renal biopsy at 4 weeks or 6 months showed signs of rejection and/or an increase in interstitial fibrosis/tubular atrophy (IF/TA). Six patients received MSC infusions. Clinical and immune monitoring was performed up to 24 weeks after MSC infusions. MSCs fulfilled the release criteria, infusions were well-tolerated, and no treatment-related serious adverse events were reported. In two recipients with allograft rejection, we had a clinical indication to perform surveillance biopsies and are able to report on the potential effects of MSCs in rejection. Although maintenance immunosuppression remained unaltered, there was a resolution of tubulitis without IF/TA in both patients. Additionally, three patients developed an opportunistic viral infection, and five of the six patients displayed a donor-specific downregulation of the peripheral blood mononuclear cell proliferation assay, not reported in patients without MSC treatment. Autologous BM MSC treatment in transplant recipients with subclinical rejection and IF/TA is clinically feasible and safe, and the findings are suggestive of systemic immunosuppression.


Assuntos
Rejeição de Enxerto/terapia , Transplante de Rim/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Adolescente , Adulto , Idoso , Células da Medula Óssea/citologia , Células Cultivadas , Feminino , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Humanos , Terapia de Imunossupressão/métodos , Rim/imunologia , Rim/patologia , Masculino , Pessoa de Meia-Idade , Transplante Autólogo , Transplante Homólogo , Resultado do Tratamento , Adulto Jovem
9.
Front Immunol ; 3: 162, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22783252

RESUMO

Ischemia/reperfusion (I/R) injury is an inevitable consequence of organ transplantation and a major determinant of patient and graft survival in kidney transplantation. Renal I/R injury can lead to fibrosis and graft failure. Although the exact sequence of events in the pathophysiology of I/R injury remains unknown, the role of inflammation has become increasingly clear. In this perspective, mesenchymal stromal cells (MSCs) are under extensive investigation as potential therapy for I/R injury, since MSCs are able to exert immune regulatory and reparative effects. Various preclinical studies indicate the beneficial effects of MSCs in ameliorating renal injury and accelerating tissue repair. These versatile cells have been shown to migrate to sites of injury and to enhance repair by paracrine mechanisms instead of by differentiating and replacing the injured cells. The first phase I studies of MSCs in human renal I/R injury and kidney transplantation have been started, and results are awaited soon. In this review, preliminary results and opportunities of MSCs in human renal I/R injury are summarized. We might be heading towards a cell-based paradigm shift in the treatment of renal I/R injury.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA