Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Publication year range
1.
Nature ; 608(7924): 778-783, 2022 08.
Article in English | MEDLINE | ID: mdl-35922516

ABSTRACT

Ferroptosis, a non-apoptotic form of cell death marked by iron-dependent lipid peroxidation1, has a key role in organ injury, degenerative disease and vulnerability of therapy-resistant cancers2. Although substantial progress has been made in understanding the molecular processes relevant to ferroptosis, additional cell-extrinsic and cell-intrinsic processes that determine cell sensitivity toward ferroptosis remain unknown. Here we show that the fully reduced forms of vitamin K-a group of naphthoquinones that includes menaquinone and phylloquinone3-confer a strong anti-ferroptotic function, in addition to the conventional function linked to blood clotting by acting as a cofactor for γ-glutamyl carboxylase. Ferroptosis suppressor protein 1 (FSP1), a NAD(P)H-ubiquinone reductase and the second mainstay of ferroptosis control after glutathione peroxidase-44,5, was found to efficiently reduce vitamin K to its hydroquinone, a potent radical-trapping antioxidant and inhibitor of (phospho)lipid peroxidation. The FSP1-mediated reduction of vitamin K was also responsible for the antidotal effect of vitamin K against warfarin poisoning. It follows that FSP1 is the enzyme mediating warfarin-resistant vitamin K reduction in the canonical vitamin K cycle6. The FSP1-dependent non-canonical vitamin K cycle can act to protect cells against detrimental lipid peroxidation and ferroptosis.


Subject(s)
Ferroptosis , Vitamin K , Antidotes/pharmacology , Antioxidants/metabolism , Antioxidants/pharmacology , Carbon-Carbon Ligases/metabolism , Coenzymes/metabolism , Ferroptosis/drug effects , Hydroquinones/metabolism , Hydroquinones/pharmacology , Lipid Peroxidation/drug effects , Oxidation-Reduction , S100 Calcium-Binding Protein A4/metabolism , Vitamin K/metabolism , Vitamin K/pharmacology , Warfarin/adverse effects
2.
Clin Transplant ; 37(3): e14880, 2023 03.
Article in English | MEDLINE | ID: mdl-36522802

ABSTRACT

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.


Subject(s)
Liver Transplantation , Humans , Bile Ducts , Bilirubin , Biomarkers , Liver , Liver Transplantation/adverse effects
3.
Eur J Immunol ; 50(12): 2041-2054, 2020 12.
Article in English | MEDLINE | ID: mdl-32640051

ABSTRACT

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, qRT2 -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-γ 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.


Subject(s)
Allografts/immunology , Antigen Presentation/immunology , Antigens, Surface/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Graft Rejection/immunology , Lectins, C-Type/immunology , Receptors, Immunologic/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
Stem Cells ; 38(6): 797-807, 2020 06.
Article in English | MEDLINE | ID: mdl-32101344

ABSTRACT

Mesenchymal stem cells (MSCs) are used in various clinical and preclinical models for immunomodulation. However, it remains unclear how the immunomodulatory effect of MSC is communicated. MSC-induced immunomodulation is known to be mediated through both MSC-secreted cytokines and direct cell-cell interactions. Recently, it has been demonstrated that metabolically inactive, heat-inactivated MSCs (HI-MSCs) have similar anti-inflammatory capacities in LPS-induced sepsis compared with viable MSC. To further investigate the immunomodulatory effects of MSC, we introduced MSC and HI-MSC in two animal models with different immunological causes. In the first model, allogeneic hearts were transplanted from C57BL/6 mice to BALB/c recipients. MSC in combination with mycophenolate mofetil (MMF) significantly improved graft survival compared with MMF alone, whereas the application of HI-MSC had no effect on graft survival. We revealed that control MSC dose-dependently inhibited CD3+ and CD8+ T-cell proliferation in vitro, whereas HI-MSC had no effect. In the second model, sepsis was induced in mice via cecal ligation and puncture. HI-MSC treatment significantly improved the overall survival, whereas control MSCs had no effect. in vitro studies demonstrated that HI-MSCs are more effectively phagocytosed by monocytes than control MSCs and induced cell death in particular of activated CD16+ monocytes, which may explain the immune protective effect of HI-MSC in the sepsis model. The results of our study demonstrate that MSC-mediated immunomodulation in sepsis is dependent on a passive recognition of MSC by monocytes, whereas fully functional MSCs are required for inhibition of T-cell-mediated allograft rejection.


Subject(s)
Heart Transplantation/methods , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Sepsis/etiology , Transplantation, Homologous/methods , Animals , Disease Models, Animal , Humans , Mice , Sepsis/pathology
5.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670793

ABSTRACT

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 TCRγ 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.


Subject(s)
Fatty Liver/etiology , Fatty Liver/immunology , Liver Transplantation/adverse effects , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Reperfusion Injury/etiology , T-Lymphocytes/immunology , Allografts/pathology , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Survival Analysis , Treatment Outcome
6.
Exp Mol Pathol ; 113: 104363, 2020 04.
Article in English | MEDLINE | ID: mdl-31881201

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is characterized by immune cell infiltration. Loss of the scaffold protein alpha-syntrophin (SNTA) protected mice from hepatic inflammation in the methionine-choline-deficient (MCD) diet model. Here, we determined increased numbers of macrophages and CD8+ T-cells in MCD diet induced NASH liver of wild type mice. In the mutant animals these NASH associated changes in immune cell composition were less pronounced. Further, there were more γδ T-cells in the NASH liver of the null mice. Galectin-3 protein in the hepatic non-parenchymal cell fraction was strongly induced in MCD diet fed wild type but not mutant mice. Antioxidant enzymes declined in NASH liver with no differences between the genotypes. To identify the target cells responsive to SNTA loss in-vitro experiments were performed. In the human hepatic stellate cell line LX-2, SNTA did not regulate pro-fibrotic or antioxidant proteins like alpha-smooth muscle actin or catalase. Soluble galectin-3 was, however, reduced upon SNTA knock-down and increased upon SNTA overexpression. SNTA deficiency neither affected cell proliferation nor cell death of LX-2 cells. In the macrophage cell line RAW264.7 low SNTA indeed caused higher galectin-3 production whereas release of TNF and cell viability were normal. Moreover, SNTA had no effect on hepatocyte chemerin and CCL2 expression. Overall, SNTA loss improved NASH without causing major effects in macrophage, hepatocyte and hepatic stellate cell lines. SNTA null mice fed the MCD diet had less body weight loss and this seems to contribute to improved liver health of the mutant mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins/deficiency , Galectin 3/metabolism , Liver/pathology , Macrophages/pathology , Membrane Proteins/deficiency , Muscle Proteins/deficiency , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/prevention & control , Actins/metabolism , Animals , Antioxidants/metabolism , Body Weight , Calcium-Binding Proteins/metabolism , Catalase/metabolism , Cell Line , Cell Proliferation , Cell Survival , Chemokine CCL2/metabolism , Chemokines/metabolism , Connective Tissue Growth Factor/metabolism , Fatty Acid Synthases/metabolism , Feeding Behavior , Heme Oxygenase-1/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-6/metabolism , L-Lactate Dehydrogenase/metabolism , Macrophages/metabolism , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Muscle Proteins/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Crit Care ; 22(1): 168, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29973233

ABSTRACT

BACKGROUND: Basic science data suggest that acute kidney injury (AKI) induced by ischemia-reperfusion injury (IRI) is an inflammatory process involving the adaptive immune response. Little is known about the T-cell contribution in the very early phase, so we investigated if tubular cellular stress expressed by elevated cell cycle biomarkers is associated with early changes in circulating T-cell subsets, applying a bedside-to-bench approach. METHODS: Our observational pilot study included 20 consecutive patients undergoing endovascular aortic repair for aortic aneurysms affecting the renal arteries, thereby requiring brief kidney hypoperfusion and reperfusion. Clinical-grade flow cytometry-based immune monitoring of peripheral immune cell populations was conducted perioperatively and linked to tubular cell stress biomarkers ([TIMP-2]•[IGFBP7]) immediately after surgery. To confirm clinical results and prove T-cell infiltration in the kidney, we simulated tubular cellular injury in an established mouse model of mild renal IRI. RESULTS: A significant correlation between tubular cell injury and a peripheral decline of γδ T cells, but no other T-cell subpopulation, was discovered within the first 24 hours (r = 0.53; p = 0.022). Turning to a mouse model of kidney warm IRI, a similar decrease in circulating γδ T cells was found and concomitantly was associated with a 6.65-fold increase in γδ T cells (p = 0.002) in the kidney tissue without alterations in other T-cell subsets, consistent with our human data. In search of a mechanistic driver of IRI, we found that the damage-associated molecule high-mobility group box 1 protein HMGB1 was significantly elevated in the peripheral blood of clinical study subjects after tubular cell injury (p = 0.019). Correspondingly, HMGB1 RNA content was significantly elevated in the murine kidney. CONCLUSIONS: Our investigation supports a hypothesis that γδ T cells are important in the very early phase of human AKI and should be considered when designing clinical trials aimed at preventing kidney damage. TRIAL REGISTRATION: ClinicalTrials.gov, NCT01915446 . Registered on 5 Aug 2013.


Subject(s)
Acute Kidney Injury/diagnosis , Acute Kidney Injury/blood , Animals , Aortic Aneurysm/blood , Aortic Aneurysm/surgery , Biomarkers/analysis , Biomarkers/blood , Disease Models, Animal , HMGB1 Protein/analysis , HMGB1 Protein/blood , Insulin-Like Growth Factor Binding Proteins/analysis , Insulin-Like Growth Factor Binding Proteins/blood , Kidney/injuries , Kidney/physiopathology , Mice, Inbred C57BL/blood , Mice, Inbred C57BL/injuries , Pilot Projects , Postoperative Complications/blood , Postoperative Complications/diagnosis , Reperfusion Injury/blood , Reperfusion Injury/diagnosis , Statistics, Nonparametric , Stress, Physiological/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tissue Inhibitor of Metalloproteinase-2/analysis , Tissue Inhibitor of Metalloproteinase-2/blood
8.
J Hepatol ; 64(1): 128-34, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26341825

ABSTRACT

BACKGROUND & AIMS: NKp46(+) cells are major effector cells in the pathogenesis of hepatic ischemia reperfusion injury (IRI). Nevertheless, the precise role of unconventional subsets like the IL-22-producing NKp46(+) cells (NK22) remains unknown. The purpose of this study was to examine the role of NK22 cells in IRI in transplantation, particularly with respect to regulation by the transcription factor ROR-gamma-t (RORγt). METHODS: To explore the role of NK22 cells in IRI in the absence of adaptive immunity, B6.RORγt-(gfp/wt)-reporter and B6.RORγt-(gfp/gfp)-knockout (KO) mice on a Rag KO background underwent 90min partial warm ischemia, followed by 24h of reperfusion. RESULTS: Rag KO mice that possess fully functional NKp46(+) cells, and Rag-common-γ-chain-double-KO (Rag-γc-DKO) mice that lack T, B and NKp46(+) cells, were used as controls. We found that Rag-γc-DKO mice lacking NK22 cells show more severe levels of hepatocellular damage (GPT, histological injury) when compared to both Rag-RORγt-reporter and Rag KO mice that possess NK22 cells. Importantly, Rag-RORγt-reporter and Rag KO mice undergoing IRI expressed high protein levels of both IL-22 and GFP (RORγt), suggesting a protective role for RORγt(+) NK22 cells in IRI. Therefore, we tested the hypothesis that RORγt critically protects from IRI through the induction of hepatic NK22 cells by studying Rag-Rorγt-DKO mice under IRI conditions. We found that the lack of RORγt(+) NK22 cells in Rag-Rorγt-DKO mice significantly enhanced IR-induced hepatocellular injury, a phenotype that could be reversed upon adoptive transfer of Rag-Rorγt-reporter NK22 cells into DKO mice. CONCLUSIONS: RORγt(+) NK22 cells play an important protective role in IRI in mice.


Subject(s)
Antigens, Ly/physiology , Interleukins/biosynthesis , Liver/blood supply , Natural Cytotoxicity Triggering Receptor 1/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/physiology , Reperfusion Injury/prevention & control , Animals , Antigens, Ly/analysis , Homeodomain Proteins/physiology , Interferon-gamma/biosynthesis , Killer Cells, Natural/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/analysis , Nuclear Receptor Subfamily 1, Group F, Member 3/analysis , Reperfusion Injury/immunology , Interleukin-22
9.
J Immunol ; 192(4): 1954-61, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24415778

ABSTRACT

We studied the developmental and functional mechanisms behind NK cell-mediated antitumor responses against metastatic colorectal carcinoma (CRC) in mice. In particular, we focused on investigating the significance of T-box transcription factors and the immunotherapeutic relevance of IL-15 in the development and function of tumor-reactive NK cells. Pulmonary CRC metastases were experimentally seeded via an adoptive i.v. transfer of luciferase-expressing CT26 CRC cells that form viewable masses via an in vivo imaging device; genetically deficient mice were used to dissect the antitumor effects of developmentally different NK cell subsets. IL-15 precomplexed to IL-15 receptor-α was used in immunotherapy experiments. We found that mice deficient for the T-box transcription factor T-bet lack terminally differentiated antitumor CD27(low)KLRG1(+) NK cells, leading to a terminal course of rapid-onset pulmonary CRC metastases. The importance of this NK cell subset for effective antitumor immunity was shown by adoptively transferring purified CD27(low)KLRG1(+) NK cells into T-bet-deficient mice and, thereby, restoring immunity against lung metastasis formation. Importantly, immunity to metastasis formation could also be restored in T-bet-deficient recipients by treating mice with IL-15 precomplexed to IL-15 receptor-α, which induced the development of eomesodermin(+)KLRG1(+) NK cells from existing NK cell populations. Thus, contingent upon their T-bet-dependent development and activation status, NK cells can control metastatic CRC in mice, which is highly relevant for the development of immunotherapeutic approaches in the clinic.


Subject(s)
Colorectal Neoplasms/pathology , Killer Cells, Natural/immunology , Lung Neoplasms/secondary , Receptors, Immunologic/metabolism , T-Box Domain Proteins/genetics , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cells, Cultured , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Homeodomain Proteins/genetics , Immunotherapy , Interferon-gamma/genetics , Interleukin-15/metabolism , Killer Cells, Natural/cytology , Lectins, C-Type , Lung Neoplasms/prevention & control , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Knockout , Perforin , Pore Forming Cytotoxic Proteins/genetics , Receptors, Interleukin-15/metabolism , Recombinant Fusion Proteins/therapeutic use , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
10.
J Immunol ; 191(1): 480-7, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23740948

ABSTRACT

An emerging body of evidence suggests a pivotal role of CD3(+) T cells in mediating early ischemia reperfusion injury (IRI). However, the precise phenotype of T cells involved and the mechanisms underlying such T cell-mediated immune responses in IRI, as well as their clinical relevance, are poorly understood. In this study, we investigated early immunological events in a model of partial warm hepatic IRI in genetically targeted mice to study the precise pathomechanistic role of RORγt(+) T cells. We found that unconventional CD27(-)γδTCR(+) and CD4(-)CD8(-) double-negative T cells are the major RORγt-expressing effector cells in hepatic IRI that play a mechanistic role by being the main source of IRI-mediating IL-17A. We further show that unconventional IRI-mediating T cells are contingent on RORγt, as highlighted by the fact that a genetic deficiency for RORγt, or its therapeutic antagonization via digoxin, is protective against hepatic IRI. Therefore, identification of CD27(-)γδTCR(+) and CD4(-)CD8(-) double-negative T cells as the major source of IL-17A via RORγt in hepatic IRI opens new therapeutic options to improve liver transplantation outcomes.


Subject(s)
Hepatitis, Animal/immunology , Hepatitis, Animal/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/physiology , Reperfusion Injury/immunology , Reperfusion Injury/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Animals , Disease Models, Animal , Genes, Reporter , Hepatitis, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Reperfusion Injury/metabolism , T-Lymphocyte Subsets/metabolism , Time Factors
11.
J Immunol ; 191(8): 4440-6, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24058178

ABSTRACT

Although Th1, Th2, and Th17 cells are thought to be major effector cells in adaptive alloimmune responses, their respective contribution to allograft rejection remains unclear. To precisely address this, we used mice genetically modified for the Th1 and Th17 hallmark transcription factors T-bet and RORγt, respectively, which allowed us to study the alloreactive role of each subset in an experimental transplant setting. We found that in a fully mismatched heterotopic mouse heart transplantation model, T cells deficient for T-bet (prone to Th17 differentiation) versus RORγt (prone to Th1 differentiation) rejected allografts at a more accelerated rate, indicating a predominance of Th17- over Th1-driven alloimmunity. Importantly, T cells doubly deficient for both T-bet and RORγt differentiated into alloreactive GATA-3-expressing Th2 cells, which promptly induced allograft rejection characterized by a Th2-type intragraft expression profile and eosinophilic infiltration. Mechanistically, Th2-mediated allograft rejection was contingent on IL-4, as its neutralization significantly prolonged allograft survival by reducing intragraft expression of Th2 effector molecules and eosinophilic allograft infiltration. Moreover, under IL-4 neutralizing conditions, alloreactive double-deficient T cells upregulated Eomesodermin (Eomes) and IFN-γ, but not GATA-3. Thus, in the absence of T-bet and RORγt, Eomes may salvage Th1-mediated alloimmunity that underlies IL-4 neutralization-resistant allograft rejection. We summarize that, whereas Th17 cells predictably promote allograft rejection, IL-4-producing GATA-3(+) Th2 cells, which are generally thought to protect allogeneic transplants, may actually be potent facilitators of organ transplant rejection in the absence of T-bet and RORγt. Moreover, Eomes may rescue Th1-mediated allograft rejection in the absence of IL-4, T-bet, and RORγt.


Subject(s)
Allografts/immunology , Graft Rejection/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Box Domain Proteins/metabolism , Th2 Cells/immunology , Adoptive Transfer , Animals , Cell Differentiation , Eosinophils/immunology , GATA3 Transcription Factor/biosynthesis , Heart Transplantation/adverse effects , Interferon-gamma/biosynthesis , Interleukin-4/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics
12.
Transplantation ; 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39294870

ABSTRACT

The outcome after liver transplantation has improved in recent years, which can be attributed to superior storage and transportation conditions of the organs, as well as better peri- and postoperative management and advancements in surgical techniques. Nevertheless, there is an increasing discrepancy between the need for organs and their availability. Consequently, the mortality rate on the waiting list is high and continues to rise. One way of counteracting this trend is to increase the use of "expanded criteria donors." This means that more and more donors will be included, especially those who are older and having additional comorbidities (eg, steatosis). A major complication of any transplantation is the occurrence of ischemia/reperfusion injury (IRI), which often leads to liver dysfunction and failure. However, there have been various promising approaches to minimize IRI in recent years, but an effective and clinically applicable method to achieve a better outcome for patients after liver transplantation is still missing. Thereby, the so-called marginal organs are predominantly affected by IRI; thus, it is crucial to develop suitable and effective treatment options for patients. Recently, regulated cell death mechanisms, particularly ferroptosis, have been implicated to play a major role in IRI, including the liver. Therefore, inhibiting this kind of cell death modality presents a promising therapeutic approach for the management of this yet untreatable condition. Thus, this review provides an overview of the role of ferroptosis in liver IRI and transplantation and discusses possible therapeutic solutions based on ferroptosis inhibition to restrain IRI in marginal organs (especially steatosis and donation after circulatory death organs).

13.
Exp Mol Pathol ; 95(2): 180-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23860432

ABSTRACT

Adiponectin receptor 1 (AdipoR1) is one of the two signaling receptors of adiponectin with multiple beneficial effects in metabolic diseases. AdipoR1 C-terminal peptide is concordant with the consensus sequence of class I PSD-95, disc large, ZO-1 (PDZ) proteins, and screening of a liver yeast two hybrid library identified binding to ß2-syntrophin (SNTB2). Hybridization of a PDZ-domain array with AdipoR1 C-terminal peptide shows association with PDZ-domains of further proteins including ß1- and α-syntrophin (SNTA). Interaction of PDZ proteins and C-terminal peptides requires a free carboxy terminus next to the PDZ-binding region and is blocked by carboxy terminal added tags. N-terminal tagged AdipoR1 is more highly expressed than C-terminal tagged receptor suggesting that the free carboxy terminus may form a complex with PDZ proteins to regulate cellular AdipoR1 levels. The C- and N-terminal tagged AdipoR1 proteins are mainly localized in the cytoplasma. N-terminal but not C-terminal tagged AdipoR1 colocalizes with syntrophins in adiponectin incubated Huh7 cells. Adiponectin induced hepatic phosphorylation of AMPK and p38 MAPK which are targets of AdipoR1 is, however, not blocked in SNTA and SNTB2 deficient mice. Further, AdipoR1 protein is similarly abundant in the liver of knock-out and wild type mice when kept on a standard chow or a high fat diet. In summary these data suggest that AdipoR1 protein levels are regulated by so far uncharacterized class I PDZ proteins which are distinct from SNTA and SNTB2.


Subject(s)
Dystrophin-Associated Proteins/metabolism , Hepatocytes/metabolism , PDZ Domains , Receptors, Adiponectin/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Cell Line , Dystrophin-Associated Proteins/chemistry , Enzyme Activation/physiology , Fluorescent Antibody Technique , Humans , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinases/metabolism , Receptors, Adiponectin/chemistry , Transfection , Two-Hybrid System Techniques , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Transl Med ; 9: 124, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21798013

ABSTRACT

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.


Subject(s)
Adult Stem Cells/transplantation , Immunomodulation , Liver Transplantation , Multipotent Stem Cells/transplantation , Stem Cell Transplantation/adverse effects , Adult , Clinical Trials Data Monitoring Committees , Feasibility Studies , Follow-Up Studies , Humans , Immunosuppressive Agents/therapeutic use , Risk Assessment
15.
J Gastroenterol Hepatol ; 25(5): 1002-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20546455

ABSTRACT

BACKGROUND: Despite pharmaceutical treatment with NTBC (2-2-nitro-4-fluoromethylbenzoyl-1,3-cyclohexanedione), a high incidence of liver malignancies occur in humans and mice suffering from hereditary tyrosinemia type 1 (HT1) caused by mutation of the fumarylacetoacetate hydrolase (fah) gene. METHODS: To evaluate the efficacy of a definitive treatment for HT1, we transfected fah knockout mice with naked plasmid DNA using high volume tail-vein injection. This approach was chosen to reduce the occurrence of insertional mutagenesis that is frequently observed when using other (retro-)viral vectors. To prolong gene expression, the fah gene was cloned between adeno-associated virus (AAV)-specific inverted terminal repeats (ITRs). RESULTS: All animals treated with high volume plasmid DNA injections could be successfully weaned off NTBC and survived in the long term without any further pharmacological support. Up to 50% fah positive hepatocytes were detected in livers of naked plasmid DNA-treated animals and serum liver function tests approximated those of wild-type controls. CONCLUSIONS: Naked plasmid DNA transfection offers a promising alternative treatment for HT1. Minimizing side-effects makes this approach especially appealing.


Subject(s)
Genetic Therapy , Hydrolases/deficiency , Liver/enzymology , Plasmids/administration & dosage , Tail/blood supply , Transfection , Tyrosinemias/therapy , Animals , Bilirubin/blood , Biomarkers/blood , Cell Proliferation , Cyclohexanones/therapeutic use , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Hepatocytes/enzymology , Hydrolases/biosynthesis , Hydrolases/genetics , Injections, Intravenous , L-Lactate Dehydrogenase/blood , Liver Function Tests , Mice , Mice, Inbred BALB C , Mice, Knockout , Nitrobenzoates/therapeutic use , Serum Albumin/metabolism , Time Factors , Tyrosinemias/drug therapy , Tyrosinemias/enzymology , Tyrosinemias/genetics , Urea/blood
16.
Liver Transpl ; 15(10): 1192-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19790154

ABSTRACT

Mesenchymal stem cells (MSCs) are promising candidate cells for immunomodulation therapy that are currently being tested in the preclinical and clinical setting. MSCs suppress the immune response in a variety of in vitro and disease models and may thus be of benefit for patients suffering from autoimmune disorders or transplant rejection. The mechanism by which MSCs modulate the immune response is still under thorough investigation, but it most likely involves expression of local factors such as indoleamine 2,3-dioxygenase, inducible nitric oxide synthase, and others as well as interactions with dendritic or antigen-presenting cells. Although MSCs have been evaluated in clinical phase I and II studies for graft-versus-host disease and heart, kidney, and bone disease, their introduction into solid organ transplantation is still eagerly awaited. In this short review, we summarize the current understanding of immunomodulation achieved by MSC therapies and introduce a possible outline for a clinical study that will use MSCs in the context of a calcineurin inhibitor-free induction protocol after liver transplantation.


Subject(s)
Immunomodulation , Liver Transplantation/methods , Mesenchymal Stem Cells/cytology , Animals , Antigen-Presenting Cells/cytology , Calcineurin Inhibitors , Coculture Techniques , Graft Rejection/immunology , Humans , Immune System , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Liver/immunology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II/metabolism
17.
In Vivo ; 23(2): 229-35, 2009.
Article in English | MEDLINE | ID: mdl-19414408

ABSTRACT

UNLABELLED: Portal vein embolization (PVE) can be used prior to liver surgery to increase the volume of the remaining liver tissue after an extensive resection. However, the application of PVE is limited and new strategies to augment liver regeneration by cellular therapy are promising alternatives. MATERIALS AND METHODS: The influence of syngeneic multipotent mesenchymal stromal cells (MSC) on liver regeneration was analysed after the ligation of the right portal vein branches in a porcine model, closely mimicking the situation of human surgery. Liver regeneration was monitored by ultrasonography, immunohistological analysis and serum biochemistry. RESULTS: The volume of the contra-lateral, non-ligated liver lobe increased in all piglets after portal vein ligation. This hyperplasia occurred earlier and was more pronounced in those piglets receiving MSC infusions as compared to non-treated controls. Biochemical liver function was stable in all pigs. Only solitary transplanted MSC were detected in recipient livers two weeks after the infusion. CONCLUSION: The infusion of porcine MSC into the portal vein in a setting of liver regeneration after surgical resection leads to accelerated and augmented hyperplasia. This effect is most likely due to bystander effects of the transplanted MSC.


Subject(s)
Embolization, Therapeutic/methods , Liver Regeneration , Mesoderm/cytology , Portal Vein/pathology , Stromal Cells/cytology , Animals , Bone Marrow Cells/cytology , Cell Proliferation , Cell Transplantation/methods , Cytokines/biosynthesis , Immunohistochemistry/methods , Liver/metabolism , Liver/pathology , Swine , Time Factors , Ultrasonography/methods
18.
Exp Hematol ; 36(11): 1507-13, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18715687

ABSTRACT

OBJECTIVE: In murine models, transplantation of wild-type bone marrow cells (BMC) can counterbalance genetic liver defects by fusion between transplanted marrow cells and resident hepatocytes. This phenomenon, however, is of no immediate clinical use because all syngeneic BMC harbor the same underlying genetic defect. MATERIALS AND METHODS: Describing the fusion between transplanted allogeneic BMC and resident hepatocytes in a murine model of hereditary tyrosinemia type I (fumarylacetoacetate hydrolase [Fah] knockout mouse), we transplanted BMC from fully allogeneic BALB/c donors into Fah(-/-) recipients after lethal total body irradiation. RESULTS: Following hematopoietic reconstitution, recipients remained healthy without pharmacological support (withdrawal of 2-2-nitro-4-fluoromethylbenzoyl-1,3-cyclohexanedione [NTBC]). Metabolic serum parameters improved nearly to wild-type levels. Livers of recipient animals contained up to 10% functional hepatocytes that stained positive for wild-type Fah, as well as both donor and recipient major histocompatibility complex. Flow cytometry confirmed this coexpression on a single cell level. Application of T-cell-depleted bone marrow reduced onset of early graft-vs-host disease. CONCLUSIONS: We introduce the observation that allogeneic bone marrow transplantation can lead to stable cell fusion of BMC with recipient hepatocytes and restored liver function in a model of otherwise lethal genetic liver disease. Thus, in principle, allogeneic cell fusion can be a possible management of hereditary liver diseases. Long-term immunological properties of fusion cells have to be further investigated.


Subject(s)
Bone Marrow Transplantation , Hydrolases/physiology , Liver/physiopathology , Animals , Hepatocytes/physiology , Histocompatibility Antigens Class I/analysis , Hydrolases/genetics , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Transplantation, Homologous
19.
Cells ; 8(11)2019 11 12.
Article in English | MEDLINE | ID: mdl-31718093

ABSTRACT

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 TNFα, were not affected by rALR. Furthermore, we evaluated infiltration of inflammatory cells into liver tissue after IRI and found no change in CD3 or γδTCR positive cells, or expression of IL17/IFNγ by γδTCR 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.


Subject(s)
Antigens, Ly/metabolism , Chemokines/genetics , Liver Regeneration/genetics , Oxidative Stress , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Animals , Apoptosis/genetics , Biopsy , Chemokines/metabolism , Disease Models, Animal , Flow Cytometry , Hepatocytes/metabolism , Immunohistochemistry , Liver/metabolism , Liver/pathology , Male , Mice , Models, Biological , Neutrophils/immunology , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL