38 research outputs found

    Augmenter of Liver Regeneration Reduces Ischemia Reperfusion Injury by Less Chemokine Expression, Gr-1 Infiltration and Oxidative Stress

    Get PDF
    Hepatic ischemia reperfusion injury (IRI) is a major complication in liver resection and transplantation. Here, we analyzed the impact of recombinant human augmenter of liver regeneration (rALR), an anti-oxidative and anti-apoptotic protein, on the deleterious process induced by ischemia reperfusion (IR). Application of rALR reduced tissue damage (necrosis), levels of lipid peroxidation (oxidative stress) and expression of anti-oxidative genes in a mouse IRI model. Damage associated molecule pattern (DAMP) and inflammatory cytokines such as HMGB1 and TNF alpha, were not affected by rALR. Furthermore, we evaluated infiltration of inflammatory cells into liver tissue after IRI and found no change in CD3 or gamma delta TCR positive cells, or expression of IL17/IFN gamma by gamma delta TCR cells. The quantity of Gr-1 positive cells (neutrophils), and therefore, myeloperoxidase activity, was lower in rALR-treated mice. Moreover, we found under hypoxic conditions attenuated ROS levels after ALR treatment in RAW264.7 cells and in primary mouse hepatocytes. Application of rALR also led to reduced expression of chemo-attractants like CXCL1, CXCL2 and CCl2 in hepatocytes. In addition, ALR expression was increased in IR mouse livers after 3 h and in biopsies from human liver transplants with minimal signs of tissue damage. Therefore, ALR attenuates IRI through reduced neutrophil tissue infiltration mediated by lower expression of key hepatic chemokines and reduction of ROS generation

    Adiponectin-induced secretion of interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1, CCL2) and interleukin-8 (IL-8, CXCL8) is impaired in monocytes from patients with type I diabetes

    Get PDF
    BACKGROUND: Systemic adiponectin is reduced in patients with cardiovascular disease (CVD) and low adiponectin may contribute to the pathogenesis of atherosclerosis. However, circulating adiponectin is elevated in type 1 diabetes (T1D) patients, who have also a higher incidence to develop CVD. Because monocytes play an important role in atherosclerosis, we analysed the influence of adiponectin on cytokine and chemokine release in monocytes from T1D patients and controls. METHODS: Systemic adiponectin was determined in the plasma and the high-molecular weight (HMW) form of adiponectin was analysed by immunoblot. Monocytes were isolated from T1D patients and controls and the adiponectin-stimulated release of interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1, CCL2) and interleukin-8 (IL-8, CXCL8) was analysed. RESULTS: Systemic adiponectin was higher in T1D patients. Immunoblot analysis of the plasma indicate abundance of HMW adiponectin in T1D patients and controls. IL-6, CCL2 and CXCL8 secretion in response to adiponectin were found induced in monocytes from controls whereas only IL-6 was upregulated in T1D cells. The induction of IL-6 by adiponectin was abrogated by an inhibitor of the NFÎşB pathway. CONCLUSION: These data indicate that adiponectin-mediated induction of IL-6, CCL2 and CXCL8 is disturbed in monocytes from T1D patients and therefore elevated systemic adiponectin in T1D patients may be less protective when compared to controls

    Analysis of organoid and immune cell co-cultures by machine learning-empowered image cytometry

    Get PDF
    Organoids are three-dimensional (3D) structures that can be derived from stem cells or adult tissue progenitor cells and exhibit an extraordinary ability to autonomously organize and resemble the cellular composition and architectural integrity of specific tissue segments. This feature makes them a useful tool for analyzing therapeutical relevant aspects, including organ development, wound healing, immune disorders and drug discovery. Most organoid models do not contain cells that mimic the neighboring tissue’s microenvironment, which could potentially hinder deeper mechanistic studies. However, to use organoid models in mechanistic studies, which would enable us to better understand pathophysiological processes, it is necessary to emulate the in situ microenvironment. This can be accomplished by incorporating selected cells of interest from neighboring tissues into the organoid culture. Nevertheless, the detection and quantification of organoids in such co-cultures remains a major technical challenge. These imaging analysis approaches would require an accurate separation of organoids from the other cell types in the co-culture. To efficiently detect and analyze 3D organoids in co-cultures, we developed a high-throughput imaging analysis platform. This method integrates automated imaging techniques and advanced image processing tools such as grayscale conversion, contrast enhancement, membrane detection and structure separation. Based on machine learning algorithms, we were able to identify and classify 3D organoids within dense co-cultures of immune cells. This procedure allows a high-throughput analysis of organoid-associated parameters such as quantity, size, and shape. Therefore, the technology has significant potential to advance contextualized research using organoid co-cultures and their potential applications in translational medicine

    Safety and feasibility of third-party multipotent adult progenitor cells for immunomodulation therapy after liver transplantation--a phase I study (MISOT-I)

    Get PDF
    BACKGROUND: Liver transplantation is the definitive treatment for many end-stage liver diseases. However, the life-long immunosuppression needed to prevent graft rejection causes clinically significant side effects. Cellular immunomodulatory therapies may allow the dose of immunosuppressive drugs to be reduced. In the current protocol, we propose to complement immunosuppressive pharmacotherapy with third-party multipotent adult progenitor cells (MAPCs), a culture-selected population of adult adherent stem cells derived from bone marrow that has been shown to display potent immunomodulatory and regenerative properties. In animal models, MAPCs reduce the need for pharmacological immunosuppression after experimental solid organ transplantation and regenerate damaged organs. METHODS: Patients enrolled in this phase I, single-arm, single-center safety and feasibility study (n=3-24) will receive 2 doses of third-party MAPCs after liver transplantation, on days 1 and 3, in addition to a calcineurin-inhibitor-free "bottom-up" immunosuppressive regimen with Basiliximab, mycophenolic acid, and steroids. The study objective is to evaluate the safety and clinical feasibility of MAPC administration in this patient cohort. The primary endpoint of the study is safety, assessed by standardized dose-limiting toxicity events. One secondary endpoint is the time until first biopsy-proven acute rejection, in order to collect first evidence of efficacy. Dose escalation (150, 300, 450, and 600 million MAPCs) will be done according to a 3 + 3 classical escalation design (4 groups of 3-6 patients each). DISCUSSION: If MAPCs are safe for patients undergoing liver transplantation in this study, a phase II/III trial will be conducted to assess their clinical efficacy

    Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion

    Get PDF
    Mesenchymal stem cells (MSC) are under investigation as a therapy for a variety of disorders. Although animal models show long term regenerative and immunomodulatory effects of MSC, the fate of MSC after infusion remains to be elucidated. In the present study the localization and viability of MSC was examined by isolation and re-culture of intravenously infused MSC. C57BL/6 MSC (500,000) constitutively expressing DsRed-fluorescent protein and radioactively labeled with Cr-51 were infused via the tail vein in wild-type C57BL/6 mice. After 5 min, 1, 24, or 72 h, mice were sacrificed and blood, lungs, liver, spleen, kidneys, and bone marrow removed. One hour after MSC infusion the majority of Cr-51 was found in the lungs, whereas after 24 h Cr-51 was mainly found in the liver. Tissue cultures demonstrated that viable donor MSC were present in the lungs up to 24 h after infusion, after which they disappeared. No viable MSC were found in the other organs examined at any time. The induction of ischemia-reperfusion injury in the liver did not trigger the migration of viable MSC to the liver. These results demonstrate that MSC are short-lived after i.v. infusion and that viable MSC do not pass the lungs. Cell debris may be transported to the liver. Long term immunomodulatory and regenerative effects of infused MSC must therefore be mediated via other cell types

    Early ÎłGT and bilirubin levels as biomarkers for regeneration and outcomes in damaged bile ducts after liver transplantation

    Get PDF
    Background Early patient and allograft survival after liver transplantation (LT) depend primarily on parenchymal function, but long-term allograft success relies often on biliary-tree function. We examined parameters related to cholangiocyte damage that predict poor long-term LT outcomes after donation after brain death (DBD). Methods Sixty bile ducts (BD) were assessed by a BD damage-score and divided into groups with “major” BD-damage (n = 33) and “no relevant” damage (n = 27) during static cold storage. Patients with “major” BD damage were further investigated by measuring biliary excretion parameters in the first 14 days post-LT (followed-up for 60-months). Results Patients who received LT showing “major” BD damage had significantly worse long-term patient survival, versus grafts with “no relevant” damage (p = .03). When “major” BD damage developed, low bilirubin levels (p = .012) and high gamma-glutamyl transferase (GGT)/bilirubin ratio (p = .0003) were evident in the early post-LT phase (7–14 days) in patients who survived (> 60 months), compared to those who did not. “High risk” patients with bile duct damage and low GGT/bilirubin ratio had significantly shorter overall survival (p < .0001). Conclusions Once “major” BD damage occurs, a high GGT/bilirubin ratio in the early post-operative phase is likely indicator of liver and cholangiocyte regeneration, and thus a harbinger of good overall outcomes. “Major” BD damage without markers of regeneration identifies LT patients that could benefit from future repair therapies

    Cross‐presentation of dead‐cell‐associated antigens by DNGR‐1⁺ dendritic cells contributes to chronic allograft rejection in mice

    Get PDF
    The purpose of this study was to elucidate whether DC NK lectin group receptor-1 (DNGR-1)-dependent cross-presentation of dead-cell-associated antigens occurs after transplantation and contributes to CD8(+)T cell responses, chronic allograft rejection (CAR), and fibrosis. BALB/c or C57BL/6 hearts were heterotopically transplanted into WT, Clec9a(-/-), or Batf3(-/-)recipient C57BL/6 mice. Allografts were analyzed for cell infiltration, CD8(+)T cell activation, fibrogenesis, and CAR using immunohistochemistry, Western blot, qRT(2)-PCR, and flow cytometry. Allografts displayed infiltration by recipient DNGR-1(+)DCs, signs of CAR, and fibrosis. Allografts in Clec9a(-/-)recipients showed reduced CAR (p < 0.0001), fibrosis (P= 0.0137), CD8(+)cell infiltration (P < 0.0001), and effector cytokine levels compared to WT recipients. Batf3-deficiency greatly reduced DNGR-1(+)DC-infiltration, CAR (P < 0.0001), and fibrosis (P= 0.0382). CD8 cells infiltrating allografts of cytochrome C treated recipients, showed reduced production of CD8 effector cytokines (P < 0.05). Further, alloreactive CD8(+)T cell response in indirect pathway IFN-gamma ELISPOT was reduced in Clec9a(-/-)recipient mice (P= 0.0283). Blockade of DNGR-1 by antibody, similar to genetic elimination of the receptor, reduced CAR (P= 0.0003), fibrosis (P= 0.0273), infiltration of CD8(+)cells (p= 0.0006), and effector cytokine levels. DNGR-1-dependent alloantigen cross-presentation by DNGR-1(+)DCs induces alloreactive CD8(+)cells that induce CAR and fibrosis. Antibody against DNGR-1 can block this process and prevent CAR and fibrosis

    Steatotic Livers Are More Susceptible to Ischemia Reperfusion Damage after Transplantation and Show Increased γδ T Cell Infiltration

    No full text
    Liver transplantation (LTx) is often the only possible therapy for many end-stage liver diseases, but successful long-term transplant outcomes are limited by multiple factors, including ischemia reperfusion injury (IRI). This situation is aggravated by a shortage of transplantable organs, thus encouraging the use of inferior quality organs. Here, we have investigated early hepatic IRI in a retrospective, exploratory, monocentric case-control study considering organ marginality. We analyzed standard LTx biopsies from 46 patients taken at the end of cold organ preparation and two hours after reperfusion, and we showed that early IRI was present after two hours in 63% of cases. Looking at our data in general, in accordance with Eurotransplant criteria, a marginal transplant was allocated at our institution in about 54% of cases. We found that patients with a marginal-organ LTx showing evidence of IRI had a significantly worse one-year survival rate (51% vs. 75%). As we saw in our study cohort, the marginality of these livers was almost entirely due to steatosis. In contrast, survival rates in patients receiving a non-marginal transplant were not influenced by the presence or absence of IRI. Poorer outcomes in marginal organs prompted us to examine pre- and post-reperfusion biopsies, and it was revealed that transplants with IRI demonstrated significantly greater T cell infiltration. Molecular analyses showed that higher mRNA expression levels of CXCL-1, CD3 and TCRÎł locus genes were found in IRI livers. We therefore conclude that the marginality of an organ, namely steatosis, exacerbates early IRI by enhancing effector immune cell infiltration. Preemptive strategies targeting immune pathways could increase the safety of using marginal organs for LTx

    Funktionsanalyse von MotE und FliK, zwei neuen Proteinen im Flagellenmotor von Sinorhizobium meliloti

    Get PDF
    Die Flagellen des Bodenbakteriums Sinorhizobium meliloti rotieren einzig im Uhrzeigersinn. Durch Variation der Rotationsgeschwindigkeit und Abstoppen einzelner Geißeln zerfällt das ehemals vorwärtstreibende Geißelbündel und die Zelle nimmt eine neue Bewegungsrichtung ein. Dieser im Vergleich zum enterobakteriellen System neuartige Mechanismus der Bewegungssteuerung bei Bakterien hat molekulare Korrelation im Vorhandensein neuer Komponenten des bakteriellen Flagellenmotors, MotC und MotE, zusätzlich zu den konservierten Protonenkanalproteinen MotA und MotB. MotC bindet an die periplasmatische Domäne des Motorproteins MotB und ist für die Motorrotation essentiell. Der erste Teil dieser Arbeit befaßt sich mit der funktionellen Charakterisierung des MotE-Proteins. Deletion von motE resultiert in Aggregation und proteolytischem Abbau des periplasmatischen MotC-Proteins, was zu Paralyse der Zellen führt. Das aus 179 Aminosäuren bestehende Polypeptid MotE wird nach der Abspaltung eines spezifischen Signalpeptides ins Periplasma exportiert, wo es über eine Disulphidbrücke zwischen den Cysteinresten 53 stabile Dimere ausbildet. Die Mutation des Cysteinrestes 53 führt zur Destabilisierung des MotE-Proteins. Stabile MotE-Dimere sind in der Lage die Aggregation von MotC-Molekülen zu inhibieren. Diese Ergebnisse lassen den Schluß zu, daß es sich bei MotE um ein spezifisches periplamsatisches Chaperon für MotC handelt. Der zweite Teil der Arbeit behandelt das Protein FliK (ehemals MotD), welches aus 475 Aminosäuren besteht und C-terminal ein konserviertes Flg_hook-Motiv besitzt. FliK-Deletionsmutanten produzieren um ein Vielfaches verlängerte Flagellenhaken, sogenannte polyhooks. Diese polyhook-Mutanten sind sowohl in der Kontrolle der Hakenlänge als auch bei der Assemblierung von Filamenten defekt. Es konnten Pseudorevertanten isoliert und charakterisiert werden, die in einer der beiden Funktionen von FliK wieder hergestellt waren. Die Mutanten zeigen nach wie vor einen Defekt bei der Kontrolle der Hakenlänge, haben aber an den polyhooks Filamente assembliert, die sich in ihrer Morphologie nicht von denen des Wildtyps unterscheiden. Die Suppressormutationen liegen alle in flhB, einem Gen, das für ein transmembranes Protein des Typ-III-Exportkanals kodiert. Basierend auf diesen Ergebnissen wird ein Modell der Hakenlängenregulation der S. meliloti Flagelle unter Einbeziehung der Rollen von FliK und FlhB vorgeschlagen

    MotD of Sinorhizobium meliloti and Related Îą-Proteobacteria Is the Flagellar-Hook-Length Regulator and Therefore Reassigned as FliK

    No full text
    The flagella of the soil bacterium Sinorhizobium meliloti differ from the enterobacterial paradigm in the complex filament structure and modulation of the flagellar rotary speed. The mode of motility control in S. meliloti has a molecular corollary in two novel periplasmic motility proteins, MotC and MotE, that are present in addition to the ubiquitous MotA/MotB energizing proton channel. A fifth motility gene is located in the mot operon downstream of the motB and motC genes. Its gene product was originally designated MotD, a cytoplasmic motility protein having an unknown function. We report here reassignment of MotD as FliK, the regulator of flagellar hook length. The FliK gene is one of the few flagellar genes not annotated in the contiguous flagellar regulon of S. meliloti. Characteristic for its class, the 475-residue FliK protein contains a conserved, compactly folded Flg hook domain in its carboxy-terminal region. Deletion of fliK leads to formation of prolonged flagellar hooks (polyhooks) with missing filament structures. Extragenic suppressor mutations all mapped in the cytoplasmic region of the transmembrane export protein FlhB and restored assembly of a flagellar filament, and thus motility, in the presence of polyhooks. The structural properties of FliK are consistent with its function as a substrate specificity switch of the flagellar export apparatus for switching from rod/hook-type substrates to filament-type substrates
    corecore