Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Carcinogenesis ; 40(2): 324-334, 2019 04 29.
Article in English | MEDLINE | ID: mdl-30698677

ABSTRACT

CD26/dipeptidyl peptidase 4 (DPP4) is a transmembrane protein which is expressed by various malignant cells. We found that the expression of CD26/DPP4 was significantly higher in lung adenocarcinoma samples in our own patient cohort compared to normal lung tissue. We therefore hypothesize that the inhibition of CD26/DPP4 can potentially suppress lung cancer growth. The CD26/DPP4 inhibitor vildagliptin was employed on Lewis Lung Carcinoma (LLC) cell line and a human lung adenocarcinoma (H460) cell line. Two weeks after subcutaneous injection of tumor cells into C57BL/6 and CD1/nude mice, the size of LLC and H460 tumors was significantly reduced by vildagliptin. Immunohistochemically, the number of macrophages (F4/80+) and NK cells (NKp46+) was significantly increased in vildagliptin-treated tumor samples. Mechanistically, we found in vitro that lung cancer cell lines expressed increased levels of surfactant protein upon vildagliptin treatment thereby promoting the pro-inflammatory activity of macrophages. By the depletion of macrophages with clodronate and by using NK cell deficient (IL-15-/-) mice, tumors reversed to the size of controls, suggesting that indeed macrophages and NK cells were responsible for the observed tumor-suppressing effect upon vildagliptin treatment. FACS analysis showed tumor-infiltrating NK cells to express tumor necrosis-related apoptosis-inducing ligand (TRAIL) which induced the intra-cellular stress marker γH2AX. Accordingly, we found upregulated γH2AX in vildagliptin-treated tumors and TRAIL-treated cell lines. Moreover, the effect of vildagliptin-mediated enhanced NK cell cytotoxicity could be reversed by antagonizing the TRAIL receptor. Our data provide evidence that the CD26/DPP4-inhibitor vildagliptin reduces lung cancer growth. We could demonstrate that this effect is exerted by surfactant-activated macrophages and NK cells that act against the tumor via TRAIL-mediated cytotoxicity.


Subject(s)
Cell Proliferation/drug effects , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Killer Cells, Natural/drug effects , Lung Neoplasms/drug therapy , Macrophages/drug effects , Vildagliptin/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Humans , Hypoglycemic Agents/pharmacology , Killer Cells, Natural/metabolism , Lung/drug effects , Lung/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , RAW 264.7 Cells
2.
Lung ; 197(2): 217-226, 2019 04.
Article in English | MEDLINE | ID: mdl-30739218

ABSTRACT

PURPOSE: Acute allograft rejection after lung transplantation remains an unsolved hurdle. The pathogenesis includes an inflammatory response during and after transplantation. Ropivacaine, an amide-linked local anesthetic, has been shown to attenuate lung injury due to its anti-inflammatory effects. We hypothesized that the drug would also be able to attenuate acute rejection (AR) after allogeneic lung transplantation. METHODS: Allogeneic, orthotopic, single left lung transplantation was performed between BALB/c (donors) and C57BL/6 (recipients) mice. Prior to explantation, lungs were flushed with normal saline with or without ropivacaine (final concentration 1 µM). Plasma levels of tumor necrosis factor-α and interleukins - 6 and - 10 were measured 3 h after transplantation by ELISA. Lung function was assessed on postoperative day five and transplanted lungs were analyzed using histology (AR), immunohistochemistry (infiltrating leukocytes) and Western blot (phosphorylation and expression of Src and caveolin-1). RESULTS: Ropivacaine pre-treatment significantly reduced AR scores (median 3 [minimum-maximum 2-4] for control vs. 2 [1-2] for ropivacaine, p < 0.001) and plasma levels of tumor necrosis factor-α (p = 0.01) compared to control, whereas plasma concentrations of interleukin - 6 (p = 0.008) and - 10 (p < 0.001) were increased by ropivacaine. The number of T-lymphocytes infiltrating the transplanted lung was attenuated (p = 0.02), while no differences in macrophage or B-lymphocyte numbers could be observed after ropivacaine pre-treatment. Caveolin-1 phosphorylation in ropivacaine-treated lungs was diminished (p = 0.004). CONCLUSIONS: Pre-treatment of donor lungs with the local anesthetic ropivacaine diminished histological signs of AR after orthotopic left lung transplantation in mice, most likely due to reduced infiltration of T-lymphocytes into the graft.


Subject(s)
Anesthetics, Local/pharmacology , Anti-Inflammatory Agents/pharmacology , Graft Rejection/prevention & control , Graft Survival/drug effects , Lung Transplantation/adverse effects , Lung/drug effects , Ropivacaine/pharmacology , Acute Disease , Allografts , Animals , Caveolin 1/metabolism , Chemotaxis, Leukocyte/drug effects , Cytokines/blood , Disease Models, Animal , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Inflammation Mediators/blood , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Phosphorylation , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors
3.
J Surg Res ; 214: 270-277, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28624055

ABSTRACT

BACKGROUND: Although sevoflurane (Sevo) had been shown to ameliorate posttransplant injury in various organs, data available are inconsistent, particularly in the context of lung transplantation (Tx). We here investigated if preconditioning by Sevo can protect from posttransplant injury regarding both, primary graft dysfunction (PGD) and acute rejection (AR) after experimental lung Tx, thereby focusing on two important clinical outcome parameters. MATERIALS AND METHODS: Three experimental approaches were used: (1) BALB/c mice were preconditioned for 2 h with Sevo or a fentanyl cocktail (Control; n = 10); (2) syngeneic (Syn) mouse lung Tx (C57BL/6) with a Sevo-preconditioned graft followed by 18 h storage to mimic PGD (Syn-Tx, n = 12) versus controls (fentanyl cocktail); and (3) allogeneic (Allo) Tx (BALB/c, donor; C57BL/6, recipient) to mimic AR (Allo-Tx, n = 12) versus controls (fentanyl cocktail). Syn-Tx grafts were harvested on Day 1, Allo-Tx grafts on Day 3 and analyzed for histology, immunohistochemistry, blood gas analysis, and inflammatory cytokines (enzyme-linked immunosorbent assay or reverse transcription polymerase chain reaction). RESULTS: Evaluating the preconditioning effect of Sevo only showed significantly better oxygenation (P = 0.03) and a tendency toward lower levels of lung tissue messenger RNA for tumor necrosis factor-α. In Syn-Tx recipients, the Sevo group had histologically a tendency toward an attenuation of PGD and showed significantly lower levels of interleukin 6 (P = 0.01) in plasma, but higher levels of interleukin 10 (P < 0.01) in lungs. Allo-Tx grafts in Sevo Tx recipients showed attenuated AR with histologically significantly lower rejection scores (P = 0.03), fewer classical macrophages (F4/80+; P < 0.01), but more anti-inflammatory activated macrophages (M2, CD206+; P < 0.01). Functionally, the Sevo group had a tendency toward improved oxygenation. CONCLUSIONS: We demonstrated that Sevo preconditioning has protective effects on lung transplants in both, PGD and AR. The observed amelioration may be attributed to suppressed inflammatory cytokines during PGD and the induction of alternatively activated macrophages during AR. These promising data could set the base for using Sevo preconditioning in donor lungs for a human trial.


Subject(s)
Graft Rejection/prevention & control , Lung Transplantation , Methyl Ethers/therapeutic use , Preoperative Care/methods , Primary Graft Dysfunction/prevention & control , Protective Agents/therapeutic use , Animals , Drug Administration Schedule , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Sevoflurane , Treatment Outcome
4.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G682-8, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26893160

ABSTRACT

Liver metastases are the most frequent cause of death due to colorectal cancer (CRC). Syngeneic orthotopic animal models, based on the grafting of cancer cells or tissue in host liver, are efficient systems for studying liver tumors and their (patho)physiological environment. Here we describe selective portal vein injection as a novel tool to generate syngeneic orthotopic models of liver tumors that avoid most of the weaknesses of existing syngeneic models. By combining portal vein injection of cancer cells with the selective clamping of distal liver lobes, tumor growth is limited to specific lobes. When applied on MC-38 CRC cells and their mouse host C57BL6, selective portal vein injection leads with 100% penetrance to MRI-detectable tumors within 1 wk, followed by a steady growth until the time of death (survival ∼7 wk) in the absence of extrahepatic disease. Similar results were obtained using CT-26 cells and their syngeneic Balb/c hosts. As a proof of principle, lobe-restricted liver tumors were also generated using Hepa1-6 (C57BL6-syngeneic) and TIB-75 (Balb/c-syngeneic) hepatocellular cancer cells, demonstrating the general applicability of selective portal vein injection for the induction of malignant liver tumors. Selective portal vein injection is technically straightforward, enables liver invasion via anatomical routes, preserves liver function, and provides unaffected liver tissue. The tumor models are reproducible and highly penetrant, with survival mainly dependent on the growth of lobe-restricted liver malignancy. These models enable biological studies and preclinical testing within short periods of time.


Subject(s)
Liver Neoplasms/pathology , Neoplasm Transplantation/methods , Animals , Cell Line , Cell Line, Tumor , Disease Models, Animal , Injections, Intravenous , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/etiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Portal Vein
5.
Hepatology ; 60(4): 1409-17, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24700614

ABSTRACT

UNLABELLED: Remote ischemic preconditioning (RIPC), the repetitive transient mechanical obstruction of vessels at a limb remote to the operative site, is a novel strategy to mitigate distant organ injury associated with surgery. In the clinic, RIPC has demonstrated efficacy in protecting various organs against ischemia reperfusion (IR), but a common mechanism underlying the systemic protection has not been identified. Here, we reasoned that protection may rely on adaptive physiological responses toward local stress, as is incurred through RIPC. Standardized mouse models of partial hepatic IR and of RIPC to the femoral vascular bundle were applied. The roles of platelets, peripheral serotonin, and circulating vascular endothelial growth factor (Vegf) were studied in thrombocytopenic mice, Tph1(-) (/) (-) mice, and through neutralizing antibodies, respectively. Models of interleukin-10 (Il10) and matrix metalloproteinase 8 (Mmp8) deficiency were used to assess downstream effectors of organ protection. The protection against hepatic IR through RIPC was dependent on platelet-derived serotonin. Downstream of serotonin, systemic protection was spread through up-regulation of circulating Vegf. Both RIPC and serotonin-Vegf induced differential gene expression in target organs, with Il10 and Mmp8 displaying consistent up-regulation across all organs investigated. Concerted inhibition of both molecules abolished the protective effects of RIPC. RIPC was able to mitigate pancreatitis, indicating that it can protect beyond ischemic insults. CONCLUSIONS: We have identified a platelet-serotonin-Vegf-Il10/Mmp8 axis that mediates the protective effects of RIPC. The systemic action, the conservation of RIPC effects among mice and humans, and the protection beyond ischemic insults suggest that the platelet-dependent axis has evolved as a preemptive response to local stress, priming the body against impending harm.


Subject(s)
Blood Platelets/physiology , Ischemic Preconditioning/methods , Liver/blood supply , Reperfusion Injury/prevention & control , Reperfusion Injury/physiopathology , Signal Transduction/physiology , Animals , Disease Models, Animal , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-10/metabolism , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Serotonin/deficiency , Serotonin/genetics , Serotonin/metabolism , Thrombocytopenia/metabolism , Thrombocytopenia/pathology , Thrombocytopenia/physiopathology , Tryptophan Hydroxylase/deficiency , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
J Hepatol ; 60(3): 625-32, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24262133

ABSTRACT

BACKGROUND & AIMS: Many of the beneficial effects of ω3-fatty acids (ω3FAs) are being attributed to their anti-inflammatory properties. In animal models, ω3FAs also protect from hepatic ischemia reperfusion injury (IRI), a significant cause of complications following liver surgery. Omegaven®, a clinical ω3FA-formulation, might counteract the exaggerated inflammatory response underlying IRI, but the according mechanisms are unresearched. Recently, GPR120 has been identified as a first receptor for ω3FAs, mediating their anti-inflammatory effects. Here, we sought to investigate whether Omegaven® protects from hepatic IRI through GPR120. METHODS: Using a mouse model of liver IRI, we compared the effects of a GPR120 agonist with those of Omegaven®. RESULTS: GPR120 in liver was located to Kupffer cells (KCs). Agonist and Omegaven® provided similar protection from IRI, which was abolished by clodronate-depletion of KCs or by pretreatment with an αGpr120-siRNA. In vitro and in vivo, both agents dampened the NFκB/JNK-mediated inflammatory response. Dampening was associated with an M1>M2 macrophage polarization shift as assessed by marker expression. In αGpr120-siRNA-pretreated mice with or without ischemia, Omegaven® was no more able to promote M2 marker expression, indicating its anti-inflammatory properties are dependent on GPR120 in liver. CONCLUSIONS: These findings establish KC-GPR120 as a key mediator of Omegaven® effects and suggest GPR120 as a therapeutic target to mitigate inflammatory stress in liver.


Subject(s)
Fish Oils/pharmacology , Kupffer Cells/physiology , Liver/drug effects , Receptors, G-Protein-Coupled/physiology , Animals , Cell Polarity , Macrophages/physiology , Mice , Signal Transduction , Triglycerides
7.
J Hepatol ; 61(2): 301-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24751831

ABSTRACT

BACKGROUND & AIMS: Fasting and calorie restriction are associated with a prolonged life span and an increased resistance to stress. The protective effects of fasting have been exploited for the mitigation of ischemic organ injury, yet the underlying mechanisms remain incompletely understood. Here, we investigated whether fasting protects liver against ischemia reperfusion (IR) through energy-preserving or anti-inflammatory mechanisms. METHODS: Fasted C57BL6 mice were subjected to partial hepatic IR. Injury was assessed by liver enzymes and histology. Raw264-7 macrophage-like cells were investigated in vitro. Sirt1 and HMGB1 were inhibited using Ex527 and neutralizing antibodies, respectively. RESULTS: Fasting for one, but not two or three days, protected from hepatic IR injury. None of the investigated energy parameters correlated with the protective effects. Instead, inflammatory responses were dampened in one-day-fasted mice and in starved macrophages. Fasting alone led to a reduction in circulating HMGB1 associated with cytoplasmic HMGB1 translocation, aggregate formation, and autophagy. Inhibition of autophagy re-elevated circulating HMGB1 and abolished protection in fasted mice, as did supplementation with HMGB1. In vitro, Sirt1 inhibition prevented HMGB1 translocation, leading to elevated HMGB1 in the supernatant. In vivo, Sirt1 inhibition abrogated the fasting-induced protection, but had no effect in the presence of neutralizing HMGB1 antibody. CONCLUSIONS: Fasting for one day protects from hepatic IR injury via Sirt1-dependent downregulation of circulating HMGB1. The reduction in serum HMGB1 appears to be mediated by its engagement in the autophagic response. These findings integrate Sirt1, HMGB1, and autophagy into a common framework that underlies the anti-inflammatory properties of short-term fasting.


Subject(s)
Fasting , HMGB1 Protein/blood , Liver/blood supply , Reperfusion Injury/prevention & control , Sirtuin 1/physiology , Animals , Down-Regulation , HMGB1 Protein/physiology , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL
8.
Gut ; 62(6): 890-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22591619

ABSTRACT

OBJECTIVE: Serotonin (5-hydroxytryptamine, 5-HT) is a potent bioactive molecule involved in a variety of physiological processes. In this study, the authors analysed whether 5-HT regulates zymogen secretion in pancreatic acinar cells and the development of pancreatic inflammation, a potentially lethal disease whose pathophysiology is not completely understood. METHODS: 5-HT regulation of zymogen secretion was analysed in pancreatic acini isolated from wild-type or tryptophan hydoxylase-1 knock-out (TPH1(-/-)) mice, which lack peripheral 5-HT, and in amylase-secreting pancreatic cell lines. Pancreatitis was induced by cerulein stimulation and biochemical and immunohistochemical methods were used to evaluate disease progression over 2 weeks. RESULTS: Absence and reduced intracellular levels of 5-HT inhibited the secretion of zymogen granules both ex vivo and in vitro and altered cytoskeleton dynamics. In addition, absence of 5-HT resulted in attenuated pro-inflammatory response after induction of pancreatitis. TPH1(-/-) mice showed limited zymogen release, reduced expression of the pro-inflammatory chemokine MCP-1 and minimal leucocyte infiltration compared with wild-type animals. Restoration of 5-HT levels in TPH1(-/-) mice recovered the blunted inflammatory processes observed during acute pancreatitis. However, cellular damage, inflammatory and fibrotic processes accelerated in TPH1(-/-) mice during disease progression. CONCLUSIONS: These results identify a 5-HT-mediated regulation of zymogen secretion in pancreatic acinar cells. In addition, they demonstrate that 5-HT is required for the onset but not for the progression of pancreatic inflammation. These findings provide novel insights into the normal physiology of pancreatic acinar cells and into the pathophysiology of pancreatitis, with potential therapeutic implications.


Subject(s)
Acinar Cells/pathology , Amylases/metabolism , Enzyme Precursors/metabolism , Pancreatitis/enzymology , Serotonin/physiology , Acinar Cells/enzymology , Actin Cytoskeleton/pathology , Animals , Cell Line , Ceruletide/adverse effects , Chemotaxis, Leukocyte , Disease Progression , Fibrosis , Immunohistochemistry , Inflammation/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis/chemically induced , Pancreatitis/pathology
9.
Am J Respir Cell Mol Biol ; 48(6): 675-84, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23392572

ABSTRACT

Despite significant progress in the treatment of chronic lung allograft rejection, obliterative bronchiolitis (OB) remains the major limitation to long-term survival after lung transplantation. The use of animal models is critical to an understanding of the pathological mechanisms behind OB, and to develop therapeutic strategies for OB. For almost 20 years, the technique of heterotopic tracheal transplantation was the leading experimental model in OB research. To address the need for a more physiologic experimental setup, a variety of small animal models have been proposed during the past two decades, such as the orthotopic tracheal transplantation model or the intrapulmonary trachea implantation model. The recent introduction of the orthotopic lung transplantation model in the mouse fulfilled the criteria for a physiologic lung transplantation setup, and also presents the advantage of being genetically modifiable. Here we review the evolution of OB models and their applications, from their beginning to the rapidly emerging physiologic models of chronic lung allograft rejection.


Subject(s)
Bronchiolitis Obliterans/pathology , Lung Transplantation/adverse effects , Respiratory Mucosa/pathology , Animals , Bronchiolitis Obliterans/immunology , Cytokines/immunology , Disease Models, Animal , Graft Rejection/immunology , Graft Rejection/pathology , Humans , Lung/immunology , Lung/metabolism , Lung/pathology , Lung Transplantation/immunology , Lung Transplantation/methods , Mice , Nitric Oxide/metabolism , Rats , Respiratory Mucosa/immunology , Trachea/metabolism , Trachea/transplantation
10.
Gastroenterology ; 143(6): 1609-1619.e4, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22960658

ABSTRACT

BACKGROUND & AIMS: Extended liver resection leads to hepatic failure because of a small remnant liver volume. Excessive parenchymal damage has been proposed as the principal cause of this failure, but little is known about the contribution of a primary deficiency in liver regeneration. We developed a mouse model to assess the regenerative capacity of a critically small liver remnant. METHODS: Extended (86%) hepatectomy (eHx) was modified to minimize collateral damage; effects were compared with those of standard (68%) partial hepatectomy (pHx) in mice. Markers of liver integrity and survival were evaluated after resection. Liver regeneration was assessed by weight gain, proliferative activity (analyses of Ki67, proliferating cell nuclear antigen, phosphorylated histone 3, mitosis, and ploidy), and regeneration-associated molecules. Knockout mice were used to study the role of p21. RESULTS: Compared with pHx, survival of mice was reduced after eHx, and associated with cholestasis and impaired liver function. However, no significant differences in hepatocyte death, sinusoidal injury, oxidative stress, or energy depletion were observed between mice after eHx or pHx. No defect in the initiation of hepatocyte proliferation was apparent. However, restoration of liver mass was delayed after eHx and associated with inadequate induction of Foxm1b and a p21-dependent delay in cell-cycle progression. In p21(-/-) mice, the cell cycle was restored, the gain in liver weight was accelerated, and survival improved after eHx. CONCLUSIONS: Significant parenchymal injury is not required for liver failure to develop after extended hepatectomy. Rather, liver dysfunction after eHx results from a transient, p21-dependent block before hepatocyte division. Therefore, a deficiency in cell-cycle progression causes liver failure after extended hepatectomy and can be overcome by inhibition of p21.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , Hepatectomy/adverse effects , Liver Failure/etiology , Liver Failure/physiopathology , Liver Regeneration/physiology , Liver/surgery , Animals , Cell Cycle/physiology , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Disease Models, Animal , Forkhead Box Protein M1 , Forkhead Transcription Factors/physiology , Liver/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/physiology
11.
Hepatology ; 56(1): 209-18, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22290718

ABSTRACT

UNLABELLED: Obstructive cholestasis induces liver injury, postoperative complications, and mortality after surgery. Adaptive control of cholestasis, including bile salt homeostasis, is necessary for recovery and survival. Peripheral serotonin is a cytoprotective neurotransmitter also associated with liver regeneration. The effect of serotonin on cholestatic liver injury is not known. Therefore, we tested whether serotonin affects the severity of cholestatic liver injury. We induced cholestasis by ligation of the bile duct (BDL) in either wild-type (WT) mice or mice lacking peripheral serotonin (Tph1(-/-) and immune thrombocytopenic [ITP] mice). Liver injury was assessed by the levels of plasma aspartate aminotransferase (AST), alanine aminotransferase (ALT) and tissue necrosis. Bile salt-regulating genes were measured by quantitative polymerase chain reaction and confirmed by western blotting and immunohistochemistry. Tph1(-/-) mice displayed higher levels of plasma AST, ALT, bile salts, and hepatic necrosis after 3 days of BDL than WT mice. Likewise, liver injury was disproportional in ITP mice. Moreover, severe cholestatic complications and mortality after prolonged BDL were increased in Tph1(-/-) mice. Despite the elevation in toxic bile salts, expression of genes involved in bile salt homeostasis and detoxification were not affected in Tph1(-/-) livers. In contrast, the bile salt reabsorption transporters Ostα and Ostß were up-regulated in the kidneys of Tph1(-/-) mice, along with a decrease in urinary bile salt excretion. Serotonin reloading of Tph1(-/-) mice reversed this phenotype, resulting in a reduction of circulating bile salts and liver injury. CONCLUSION: We propose a physiological function of serotonin is to ameliorate liver injury and stabilize the bile salt pool through adaptation of renal transporters in cholestasis.


Subject(s)
Bile Acids and Salts/metabolism , Liver Diseases/prevention & control , Liver/immunology , Serotonin/metabolism , Animals , Bile Ducts/surgery , Cells, Cultured , Cholestasis/metabolism , Cholestasis/pathology , Disease Models, Animal , Hepatocytes/cytology , Hepatocytes/metabolism , Ligation/methods , Liver/pathology , Liver Diseases/blood , Liver Function Tests , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Sensitivity and Specificity , Serotonin/pharmacology
12.
J Surg Res ; 185(1): 426-32, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23890404

ABSTRACT

BACKGROUND: Mouse lung transplantation has evolved into an established scientific model that is currently used by an increasing number of research groups. Acquiring this technique without previous microsurgical knowledge is considered very difficult. Disclosing all the intraoperative failures and mistakes during the model's evolution will encourage all researchers who lack microsurgical skills that overcoming and eventually succeeding in this model is possible. METHODS: Inbred (C57BL/6, BALB/c, SVG129) and CD1-outbred mice served as the transplant donors and recipients. The training procedure was performed by a surgeon not experienced in microsurgery, and arranged as follows: donor preparation until proof of functionality, graft implantation into deceased recipients, and graft implantation into surviving recipients until stable performance was achieved. The transplant's viability was controlled using micro-computed tomography imaging. RESULTS: Donor preparation complications decreased from 43% after 1 month to 0% after 2 mo. The first functional donor was implanted at day 28, and the first successful implantation into a surviving recipient was performed at day 60 after six training recipients. Micro-computed tomography confirmed a ventilated and perfused graft. Intraoperative complications, mainly due to anastomosis failure, decreased from 58% after the first month to 15% at the latest assessment. The most recent implantation time was 75 ± 4.8 min, and the transplantation success rate was 82% ± 2.8%. A modified forceps considerably improved completion of the venous anastomosis. CONCLUSIONS: Consistent success in the mouse lung transplantation model can be achieved even without pre-existing microsurgical skills. The surgery can be mastered within a reasonable period using a limited number of training animals. Procedure-related complications can be restricted to a minimum by applying key corrective steps at critical phases. This should encourage investigators without pre-expert knowledge in microsurgery to start to learn this research model.


Subject(s)
Graft Survival , Lung Transplantation/education , Lung Transplantation/methods , Microsurgery/education , Microsurgery/methods , Anastomosis, Surgical/education , Anastomosis, Surgical/methods , Animals , Animals, Outbred Strains , Intraoperative Complications/prevention & control , Lung/diagnostic imaging , Lung/surgery , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Operative Time , Postoperative Complications/prevention & control , Specific Pathogen-Free Organisms , X-Ray Microtomography
13.
Ann Surg ; 256(5): 730-7; discussion 737-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23095616

ABSTRACT

BACKGROUND: The aim of hyperthermic intraperitoneal chemotherapy (HIPEC) is to eradicate microscopic residual tumor after radical surgical tumor excision in patients with peritoneal carcinomatosis. The common use of antineoplastic agents such as mitomycin C, doxorubicin, or oxaliplatin with hyperthermia fails to eradicate tumors in a significant subset of patients, and alternative approaches to target chemoresistant cells are needed. The induction of reactive oxygen species (ROS) by inhibiting the critical detoxification enzyme superoxide dismutase (SOD) during hyperthermia is an appealing approach to induce death of residual cancer cells. METHODS: Human and murine colon cancer cell lines were subjected to mild hyperthermia (40-42°C), and treated with chemotherapy, similar to clinical protocols. ROS were induced by the SOD inhibitor diethyldithiocarbamate (DDC), a metabolite of the drug disulfiram. In mice, peritoneal carcinomatosis use C57Bl/6 was induced in C57Bl/6 by intraperitoneal injection of syngenic tumor cells (MC38). RESULTS: Hyperthermia alone failed to kill cells but induced intracellular ROS and activated protective mechanisms. Chemotherapy conferred inconsistent cytotoxicity depending on the cell line and dose. In contrast, induction of ROS by DDC consistently activated apoptotic pathways, with increased cell death in combination with mild hyperthermia. In vivo, combined treatment with DDC and hyperthermia significantly delayed tumor progression in tumor-bearing mice. In addition, hyperthermic combined treatment with chemotherapy and DDC significantly improved animal survival compared with chemotherapy alone. CONCLUSIONS: Addition of DDC improves the efficacy of existing HIPEC protocols in a safe way and may open the door to a more effective, multimodal HIPEC.


Subject(s)
Antineoplastic Agents/administration & dosage , Chemotherapy, Cancer, Regional Perfusion , Colonic Neoplasms/drug therapy , Hyperthermia, Induced , Mitomycin/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Ditiocarb , Dose-Response Relationship, Drug , Humans , Hydrogen Peroxide/pharmacology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mitomycin/pharmacology , Neoplasms, Experimental/drug therapy , Oxidative Stress , Peritoneal Cavity , Reactive Oxygen Species/pharmacology
14.
J Biomater Appl ; 36(10): 1826-1837, 2022 05.
Article in English | MEDLINE | ID: mdl-35285347

ABSTRACT

Chest wall repair can be necessary after tumor resection or chest injury. In order to cover or replace chest wall defects, autologous tissue or different synthetic materials are commonly used, among them the semi-rigid gold standard Gore-Tex® and prolene meshes. Synthetic tissues include composite materials with an organic and an inorganic component. On the basis of previously reported hybrid nanocomposite poly-lactic-co-glycolic acid amorphous calcium phosphate nanocomposite (PLGA/aCaP), a CuO component was incorporated to yield (60%/35%/5%). This graft was tested in vitro by seeding with murine adipose-derived stem cells (ASCs) for cell attachment and migration. The graft was compared to PLGA/CaCO3 and PLGA/hydroxyapatite, each providing the inorganic phase as nanoparticles. Further characterization of the graft was performed using scanning electron microscopy. Furthermore, PLGA/aCaP/CuO was implanted as a chest wall graft in mice. After 4 weeks, total cell density, graft integration, extracellular matrix components such as fibronectin and collagen I, the cellular inflammatory response (macrophages, F4/80 and lymphocytes, CD3) as well as vascularization (CD31) were quantitatively assessed. The nanocomposite PLGA/aCaP/CuO showed a good cell attachment and cells migrated well into the pores of the electrospun meshes. Cell densities did not differ between PLGA/aCaP/CuO and PLGA/CaCO3 or PLGA/hydroxyapatite, respectively. When applied as a chest wall graft, adequate stability for suturing into the thoracic wall could be achieved. Four weeks post-implantation, there was an excellent tissue integration without relevant fibrotic changes and a predominating collagen I matrix deposition within the graft. Slightly increased inflammation, reflected by increased infiltration of macrophages could be observed. Vascularization of the graft was significantly enhanced when compared with PLGA/aCaP (no CuO). We conclude that the hybrid nanocomposite PLGA/aCaP/CuO is a viable option to be used as a chest wall graft. Surgical implantation of the material is feasible and provides stability and enough flexibility. Proper tissue integration and an excellent vascularization are characteristics of this biodegradable material.


Subject(s)
Nanocomposites , Nanoparticles , Thoracic Wall , Animals , Copper , Mice , Oxides , Thoracic Wall/surgery , Tissue Engineering , Tissue Scaffolds
15.
Cancers (Basel) ; 14(13)2022 Jun 25.
Article in English | MEDLINE | ID: mdl-35804886

ABSTRACT

Tumor grafts grown on the chorioallantoic membrane (CAM) of chicken embryos represent a transition between cell culture and mammalian in vivo models. Magnetic resonance imaging (MRI) started to harness this potential. Functional gas challenge is feasible on the CAM. Using quantitative T1 and T2* mapping, we characterized the response of MC-38 colon, A549, and H460 adeno-carcinoma cell grafts to hypercapnic (HC) and hypercapnic-hyperoxic (HCHO) gas challenges, pertaining to the grafts' vascular and oxygenation phenotypes. MR imaging revealed that larger T1 and T2* were located in the center of H460 and MC-38 tumors. Quantitative analysis showed a significant reduction in T1 and a significant increase in T2* in response to HCHO for A549 grafts, while H460 and MC-38 tumors did not respond to either gas challenge. Different tumor grafts respond differentially to HC and HCHO conditions. A549 tumor grafts, with higher vessel density and smaller tumor diameter compared with H460 and MC-38 grafts, had a significant response in T1 for HCHO and T2* increased slightly during HC and significantly under HCHO, consistent with a normoxic phenotype and functional vasoreactivity. Therefore, gas challenges enable differential characterization of tumor grafts with respect to their vascular and oxygenation status.

16.
Cancers (Basel) ; 14(20)2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36291910

ABSTRACT

Human lung cancer ranks among the most frequently treated cancers worldwide. As copper appears critical to angiogenesis and tumor growth, selective removal of copper represents a promising strategy to restrict tumor growth. To this end, we explored the activity of the novel high-affinity membrane-permeant Cu(I) chelator PSP-2 featuring a low-zeptomolar dissociation constant. Using H460 human lung cancer cells, we generated small tumors on the chorioallantoic membrane of the chicken embryo (CAM assay) and studied the effects of topical PSP-2 application on their weight and vessel density after one week. We observed a significant angiosuppression along with a marked decrease in tumor weight under PSP-2 application compared to controls. Moreover, PSP-2 exposure resulted in lower ki67+ cell numbers at a low dose but increased cell count under a high dose. Moreover, HIF-1α+ cells were significantly reduced with low-dose PSP-2 exposure compared to high-dose and control. The total copper content was considerably lower in PSP-2 treated tumors, although statistically not significant. Altogether, PSP-2 shows promising potential as an anti-cancer drug. Nevertheless, further animal experiments and application to different tumor types are mandatory to support these initial findings, paving the way toward clinical trials.

17.
J Hepatol ; 55(6): 1291-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21703192

ABSTRACT

BACKGROUND & AIMS: Chemical composition of hepatic lipids is an evolving player in steatotic liver ischemia/reperfusion (I/R) injury. Thromboxane A(2) (TXA(2)) is a vasoactive pro-inflammatory lipid mediator derived from arachidonic acid (AA), an omega-6 fatty acid (Ω-6 FA). Reduced tolerance of the macrosteatotic liver to I/R may be related to increased TXA(2) synthesis due to the predominance of Ω-6 FAs. METHODS: TXA(2) levels elicited by I/R in ob/ob and wild type mice were assessed by ELISA. Ob/ob mice were fed Ω-3 FAs enriched diet to reduce hepatic synthesis of AA and TXA(2) or treated with selective TXA(2) receptor blocker before I/R. RESULTS: I/R triggered significantly higher hepatic TXA(2) production in ob/ob than wild type animals. Compared with ob/ob mice on regular diet, Ω-3 FAs supplementation markedly reduced hepatic AA levels before ischemia and consistently blunted hepatic TXA(2) synthesis after reperfusion. Sinusoidal perfusion and hepatocellular damage were significantly ameliorated despite downregulation of heme oxygenase-1. Hepatic transcript and protein levels of IL-1ß and neutrophil recruitment were significantly diminished after reperfusion. Moreover, TXA(2) receptor blockage conferred similar protection without modification of the histological pattern of steatosis. A stronger protection was achieved in the steatotic compared with lean animals. CONCLUSIONS: Enhanced I/R injury in the macrosteatotic liver is explained, at least partially, by TXA(2) mediated microcirculatory failure rather than size-related mechanical compression of the sinusoids by lipid droplets. TXA(2) blockage may be a simple strategy to include steatotic organs and overcome the shortage of donor organs for liver transplantation.


Subject(s)
Fatty Liver/metabolism , Lipids/chemistry , Liver/injuries , Liver/metabolism , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Thromboxane A2/metabolism , 6-Ketoprostaglandin F1 alpha/biosynthesis , Animals , Arachidonic Acid/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Liver/complications , Fatty Liver/pathology , Lipid Metabolism , Liver/blood supply , Liver/drug effects , Macrophage Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Microcirculation/drug effects , Neutrophil Activation/drug effects , Oxidative Stress/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Reperfusion Injury/pathology
18.
Hepatology ; 51(4): 1244-54, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20099302

ABSTRACT

UNLABELLED: In addition to its function as a neurotransmitter and vascular active molecule, serotonin is also a mitogen for hepatocytes and promotes liver regeneration. A possible role in hepatocellular cancer has not yet been investigated. Human hepatocellular cancer cell lines Huh7 and HepG2 were used to assess the function of serotonin in these cell lines. Characteristics of autophagy were detected with transmission electron microscopy, immunoblots of microtubule-associated protein light chain 3(LC3) and p62 (sequestosome 1). Immunoblots of the mammalian target of rapamycin (mTOR) and its downstream targets p70S6K and 4E-BP1 were used to investigate signaling pathways of serotonin. Two different animal models served as principle of proof of in vitro findings. Clinical relevance of the experimental findings was evaluated with a tissue microarray from 168 patients with hepatocellular carcinoma. Serotonin promotes tumor growth and survival in starved hepatocellular carcinoma cells. During starvation hepatocellular carcinoma cells exhibited characteristics of autophagy, which disappeared in serotonin-treated cells. Rapamycin, an inhibitor of mTOR, is known to induce autophagy. Serotonin could override rapamycin by an mTOR-independent pathway and activate common downstream signals such as p70S6K and 4E-BP1. In two tumor models of the mouse, inhibition of serotonin signaling consistently impaired tumor growth. Human biopsies revealed expression of the serotonin receptor HTR2B, correlating with downstream signals, e.g., phosphorylated p70S6K and proliferation. CONCLUSION: This study provides evidence that serotonin is involved in tumor growth of hepatocellular cancer by activating downstream targets of mTOR, and therefore serotonin-related pathways might represent a new treatment strategy.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Serotonin/pharmacology , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytoprotection , Disease Models, Animal , Female , Humans , Intracellular Signaling Peptides and Proteins/physiology , Mice , Protein Serine-Threonine Kinases/physiology , Receptor, Serotonin, 5-HT2B/physiology , Signal Transduction , TOR Serine-Threonine Kinases
19.
Liver Int ; 31(3): 313-21, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21281431

ABSTRACT

BACKGROUND: New chemotherapy regimens are increasingly used in metastatic colorectal cancer to the liver before surgery. Some clinical observations have suggested that chemotherapy may affect liver regeneration. AIMS: The aim of this study was to evaluate liver damage and liver regeneration after chemotherapy treatment in a model of partial hepatectomy. METHODS: C57BL/6 mice were repeatedly treated with intraperitoneal injections of either saline or different chemotherapy regimens including the drugs 5-fluorouracyl (5-FU), irinotecan, oxaliplatin, gemcitabine and combined treatments with 5-FU/irinotecan, 5-FU/oxaliplatin. A 70% partial hepatectomy was performed 1 week after the last injection. Ki-67 and PCNA immunohistochemistry were performed to assess liver regeneration, serum liver enzymes and histology analysis to evaluate injury. RESULTS: A variety of chemotherapeutic agents used at maximum tolerated doses compatible with survival affected body weight and blood cell levels. However, these regimens did not affect liver injury before and after hepatectomy nor did they impair liver regeneration. Liver histology showed no steatosis, fibrosis or inflammation before hepatectomy. We therefore tested whether chemotherapy in presence of diet-induced steatosis may trigger injury. Even under these conditions, we did not observe histological signs of inflammation or sinusoidal injury. CONCLUSIONS: Liver injury and liver regeneration are not impaired after neoadjuvant chemotherapy with 5-FU, irinotecan, oxaliplatin and gemcitabine in non-tumoural liver parenchyma. In addition, combined treatments disclose no adverse effects on liver regeneration. Chemotherapy alone induces no histological alterations even in the presence of steatosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/toxicity , Chemical and Drug Induced Liver Injury/etiology , Disease Models, Animal , Hepatectomy , Liver Regeneration/drug effects , Liver/drug effects , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Fatty Liver/complications , Fatty Liver/pathology , Ki-67 Antigen/metabolism , Liver/metabolism , Liver/pathology , Liver Regeneration/physiology , Mice , Mice, Inbred C57BL , Proliferating Cell Nuclear Antigen/metabolism
20.
Life Sci ; 278: 119496, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-33894269

ABSTRACT

AIMS: Chronic lung allograft dysfunction (CLAD) after lung transplantation (Tx) is the clinical result of chronic airway rejection lesions (CARL), histomorphologically described as either obliterative remodeling of small airways or alveolar fibroelastosis, or as a combination of both. We here investigated the CD26-inhibitory effect on CD26-expressing CARL. MAIN METHODS: CARL were induced by BALB/c â†’ C57BL/6 mouse Tx under mild immunosuppression. CARL-related pro-fibrotic mediators were determined by RT-qPCR and western blotting (WB), EMT and ERK markers by WB. CD26 co-expression by immunofluorescence. CD26 was inhibited by Vildagliptin, gene depleted by CD26-/- mice. Primary lung fibroblasts were employed for ex vivo analyses. Samples from lung transplant patients with CLAD were analyzed by immunohistochemistry. KEY FINDINGS: CARL revealed a significantly higher expression of profibrotic proteins vs. normal lungs (p < 0.05). CD26 and EMT co-expressed in CARL with significantly higher Vimentin, Slug, Hif-1α, α-SMA expression vs. normal lungs (p < 0.05). Vildagliptin decreased the expression of α-SMA and N-cadherin in wild type (WT) lung fibroblasts (p < 0.05). Primary lung fibroblasts from WT and CD26-/- mice treated with TGF-ß1, IFN-γ, and FGF showed a reduction of EMT protein expression, proliferation, and reduced activation of ERK in CD26-/- mice vs. WT mice. CD26-positive cells were found in patient samples with CLAD in areas of loose fibrosis, but not in areas of dense fibrosis. SIGNIFICANCE: CD26 is expressed in CARL-developing lung transplants and CD26-inhibition downregulates fibrosis-forming mediators and fibroblast proliferation. CD26 thus qualifies as a target to attenuate the development of CARL mainly via modulation of ERK and the EMT pathway.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Lung Diseases/physiopathology , Actins/metabolism , Animals , Cadherins/metabolism , Chronic Disease , Fibroblasts/metabolism , Fibrosis/pathology , Graft Rejection , Immunosuppression Therapy , Lung/metabolism , Lung Transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , Primary Graft Dysfunction , Vildagliptin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL