Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Int J Mol Sci ; 23(16)2022 Aug 18.
Article in English | MEDLINE | ID: mdl-36012587

ABSTRACT

The biomedical consequences of allogeneic blood transfusions and the possible pathomechanisms of transfusion-related morbidity and mortality are still not entirely understood. In retrospective studies, allogeneic transfusion was associated with increased rates of cancer recurrence, metastasis and death in patients with colorectal cancer. However, correlation does not imply causation. The purpose of this study was to elucidate this empirical observation further in order to address insecurity among patients and clinicians. We focused on the in vitro effect of microparticles derived from red blood cell units (RMPs). We incubated different colon carcinoma cells with RMPs and analyzed their effects on growth, invasion, migration and tumor marker expression. Furthermore, effects on Wnt, Akt and ERK signaling were explored. Our results show RMPs do not seem to affect functional and phenotypic characteristics of different colon carcinoma cells and did not induce or inhibit Wnt, Akt or ERK signaling, albeit in cell culture models lacking tumor microenvironment. Allogeneic blood transfusions are associated with poor prognosis, but RMPs do not seem to convey tumor-enhancing effects. Most likely, the circumstances that necessitate the transfusion, such as preoperative anemia, tumor stage, perioperative blood loss and extension of surgery, take center stage.


Subject(s)
Carcinoma , Cell-Derived Microparticles , Colonic Neoplasms , Carcinoma/complications , Cell-Derived Microparticles/pathology , Colonic Neoplasms/pathology , Humans , Neoplasm Recurrence, Local/etiology , Proto-Oncogene Proteins c-akt , Retrospective Studies , Tumor Microenvironment
2.
Int J Mol Sci ; 21(6)2020 Mar 16.
Article in English | MEDLINE | ID: mdl-32188161

ABSTRACT

Lipocalin-2 (Lcn-2) is rapidly upregulated in macrophages after renal tubular injury and acts as renoprotective and pro-regenerative agent. Lcn-2 possesses the ability to bind and transport iron with high affinity. Therefore, the present study focuses on the decisive role of the Lcn-2 iron-load for its pro-regenerative function. Primary mouse tubular epithelial cells were isolated from kidney tissue of wildtype mice and incubated with 5µM Cisplatin for 24h to induce injury. Bone marrow-derived macrophages of wildtype and Lcn-2-/- mice were isolated and polarized with IL-10 towards an anti-inflammatory, iron-release phenotype. Their supernatants as well as recombinant iron-loaded holo-Lcn-2 was used for stimulation of Cisplatin-injured tubular epithelial cells. Incubation of tubular epithelial cells with wildtype supernatants resulted in less damage and induced cellular proliferation, whereas in absence of Lcn-2 no protective effect was observed. Epithelial integrity as well as cellular proliferation showed a clear protection upon rescue experiments applying holo-Lcn-2. Notably, we detected a positive correlation between total iron amounts in tubular epithelial cells and cellular proliferation, which, in turn, reinforced the assumed link between availability of Lcn-2-bound iron and recovery. We hypothesize that macrophage-released Lcn-2-bound iron is provided to tubular epithelial cells during toxic cell damage, whereby injury is limited and recovery is favored.


Subject(s)
Epithelial Cells/metabolism , Kidney/metabolism , Lipocalin-2/metabolism , Macrophages/metabolism , Regeneration , Animals , Cell Proliferation , Cisplatin/adverse effects , Iron/metabolism , Kidney/drug effects , Kidney/injuries , Lipocalin-2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins , Up-Regulation
3.
Cancers (Basel) ; 12(3)2020 Feb 25.
Article in English | MEDLINE | ID: mdl-32106629

ABSTRACT

Accumulating evidence suggests that iron homeostasis is disturbed in tumors. We aimed at clarifying the distribution of iron in renal cell carcinoma (RCC). Considering the pivotal role of macrophages for iron homeostasis and their association with poor clinical outcome, we investigated the role of macrophage-secreted iron for tumor progression by applying a novel chelation approach. We applied flow cytometry and multiplex-immunohistochemistry to detect iron-dependent markers and analyzed iron distribution with atomic absorption spectrometry in patients diagnosed with RCC. We further analyzed the functional significance of iron by applying a novel extracellular chelator using RCC cell lines as well as patient-derived primary cells. The expression of iron-regulated genes was significantly elevated in tumors compared to adjacent healthy tissue. Iron retention was detected in tumor cells, whereas tumor-associated macrophages showed an iron-release phenotype accompanied by enhanced expression of ferroportin. We found increased iron amounts in extracellular fluids, which in turn stimulated tumor cell proliferation and migration. In vitro, macrophage-derived iron showed pro-tumor functions, whereas application of an extracellular chelator blocked these effects. Our study provides new insights in iron distribution and iron-handling in RCC. Chelators that specifically scavenge iron in the extracellular space confirmed the importance of macrophage-secreted iron in promoting tumor growth.

4.
Br J Cancer ; 122(3): 421-433, 2020 02.
Article in English | MEDLINE | ID: mdl-31772326

ABSTRACT

BACKGROUND: We aimed at clarifying the role of lipocalin-2 (LCN-2) in clear-cell renal cell carcinoma (ccRCC). Since LCN-2 was recently identified as a novel iron transporter, we explored its iron load as a decisive factor in conferring its biological function. METHODS: LCN-2 expression was analysed at the mRNA and protein level by using immunohistochemistry, RNAscope® and qRT-PCR in patients diagnosed with clear-cell renal cell carcinoma compared with adjacent healthy tissue. We measured LCN-2-bound iron by atomic absorption spectrometry from patient-derived samples and applied functional assays by using ccRCC cell lines, primary cells, and 3D tumour spheroids to verify the role of the LCN-2 iron load in tumour progression. RESULTS: LCN-2 was associated with poor patient survival and LCN-2 mRNA clustered in high- and low-expressing ccRCC patients. LCN-2 protein was found overexpressed in tumour compared with adjacent healthy tissue, whereby LCN-2 was iron loaded. In vitro, the iron load determines the biological function of LCN-2. Iron-loaded LCN-2 showed pro-tumour functions, whereas iron-free LCN-2 produced adverse effects. CONCLUSIONS: We provide new insights into the pro-tumour function of LCN-2. LCN-2 donates iron to cells to promote migration and matrix adhesion. Since the iron load of LCN-2 determines its pro-tumour characteristics, targeting either its iron load or its receptor interaction might represent new therapeutic options.


Subject(s)
Carcinoma, Renal Cell/metabolism , Iron/metabolism , Kidney Neoplasms/metabolism , Lipocalin-2/metabolism , RNA, Messenger/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/surgery , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Female , Humans , In Vitro Techniques , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Lipocalin-2/genetics , Male , Middle Aged , Prognosis , Real-Time Polymerase Chain Reaction , Spectrophotometry, Atomic , Spheroids, Cellular , Tumor Cells, Cultured
5.
Cell Tissue Res ; 378(1): 143-154, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30989399

ABSTRACT

It is well known that sepsis and inflammation reduce male fertility. Within the testis, toll-like receptor 3 (TLR3) is constitutively expressed and recognizes double-stranded RNA (dsRNA) from viruses, degraded bacteria, damaged tissues and necrotic cells. To characterize the potential role of TLR3 in response to testicular infections, its expression and downstream signaling were investigated upon challenge with lipopolysaccharides (LPS) in two mouse strains that differ in their immuno-competence regarding T cell-regulated immunity. Thereto, Balb/c and Foxn1nu mice were randomized into six interventional groups treated with either i.v. application of saline or LPS followed by 20 min, 5 h 30 min and 18 h of observation and two sham-treated control groups. LPS administration induced a significant stress response; the amplification was manifested for TLR3 and interleukin 6 (IL6) mRNA in the impaired testis 5 h 30 min after LPS injection. TLR3 immunostaining revealed that TLR3 was primarily localized in spermatocytes. The TLR3 expression displayed different temporal dynamics between both mouse strains. However, immunofluorescence staining indicated only punctual interferon regulatory factor 3 (IRF3) expression upon LPS treatment along with minor alterations in interferon ß (IFNß) mRNA expression. Induction of acute inflammation was closely followed by a significant shift of the Bax/Bcl2 ratio to pro-apoptotic signaling accompanied by augmented TUNEL-positive cells 18 h after LPS injection with again differing patterns in both mouse strains. In conclusion, this study shows the involvement of TLR3 in response to LPS-induced testicular inflammation in immuno-competent and -incompetent mice, yet lacking transmission into its signaling pathway.


Subject(s)
Apoptosis/immunology , Orchitis/immunology , Spermatocytes/immunology , Testis/metabolism , Toll-Like Receptor 3/immunology , Animals , Interferon Regulatory Factor-3/immunology , Interferon-beta/immunology , Lipopolysaccharides/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Orchitis/chemically induced , Spermatocytes/cytology , Testis/pathology
6.
J Cancer Res Clin Oncol ; 145(6): 1405-1416, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30903318

ABSTRACT

PURPOSE: NOD1 and NOD2 (nucleotide-binding oligomerization domain)-receptors are intracellular receptors and belong to the family of pattern recognition receptors being present in both human and murine renal tubular cells. Besides, NOD1 has been proved to promote apoptosis, upon its overexpression. Hence, we aimed to investigate NOD1 and NOD2 expression in human clear cell renal cell carcinoma (ccRCC). METHODS: Tumor and corresponding adjacent healthy tissues from 41 patients with histopathological diagnosis of ccRCC as well as primary isolated renal tubular epithelial cells (TECs) and tumor tissue from a murine xenograft model using CAKI-1 ccRCC cells were analyzed. RESULTS: NOD1 and NOD2 mRNA was constitutively expressed in both tumor and adjacent healthy renal tissue, with NOD1 being significantly lower and in contrast NOD2 significantly higher expressed in tumor tissue compared to healthy tissues. Immunohistochemically, NOD1 was located not only in the cytoplasm, but also in the nucleus in ccRCC tissue whereas NOD2 was solely localized in the cytoplasm in both human ccRCC as well as in the healthy tubular system. Focusing on the vasculature, NOD2 displayed broader expression than NOD1. In primary TECs as well as CAKI-1 cells NOD1 and NOD2 was constitutively expressed and increasable upon LPS stimulation. In the mouse xenograft model, human NOD1 mRNA was significantly higher expressed compared to NOD2. In contrast hereto, we observed a shift towards lower mouse NOD1 compared to NOD2 mRNA expression. CONCLUSION: In view of reduced apoptosis-associated NOD1 expression in ccRCC tissue opposed to higher expression of NOD2 in tumor vasculature, inducibility of NOD expression in TECs as well as the detected shift of NOD1 and NOD2 expression in the mouse xenograft model, modulation of NOD receptors might, therefore, provide a molecular therapeutic approach in ccRCC.


Subject(s)
Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Heterografts , Humans , Immunity, Innate , Immunohistochemistry , Kidney/blood supply , Kidney/immunology , Kidney Neoplasms/blood supply , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Tubules/immunology , Male , Mice , Middle Aged , Nod1 Signaling Adaptor Protein/biosynthesis , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/biosynthesis , Nod2 Signaling Adaptor Protein/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Int J Mol Sci ; 19(6)2018 06 10.
Article in English | MEDLINE | ID: mdl-29890767

ABSTRACT

Stem cell-based therapies require cells with a maximum regenerative capacity in order to support regeneration after tissue injury and organ failure. Optimization of this regenerative potential of mesenchymal stromal/stem cells (MSC) or their conditioned medium by in vitro preconditioning regimens are considered to be a promising strategy to improve the release of regenerative factors. In the present study, MSC were isolated from inguinal adipose tissue (mASC) from C57BL/6 mice, cultured, and characterized. Then, mASC were either preconditioned by incubation in a hypoxic environment (0.5% O2), or in normoxia in the presence of murine epidermal growth factor (EGF) or tumor necrosis factor α (TNFα) for 48 h. Protein expression was measured by a commercially available array. Selected factors were verified by PCR analysis. The expression of 83 out of 308 proteins (26.9%) assayed was found to be increased after preconditioning with TNFα, whereas the expression of 61 (19.8%) and 70 (22.7%) proteins was increased after incubation with EGF or in hypoxia, respectively. Furthermore, we showed the proliferation-promoting effects of the preconditioned culture supernatants on injured epithelial cells in vitro. Our findings indicate that each preconditioning regimen tested induced an individual expression profile with a wide variety of factors, including several growth factors and cytokines, and therefore may enhance the regenerative potential of mASC for cell-based therapies.


Subject(s)
Adipose Tissue/cytology , Cell Culture Techniques/methods , Gene Expression Profiling , Mesenchymal Stem Cells/cytology , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Expression Regulation/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Biochem Pharmacol ; 155: 48-60, 2018 09.
Article in English | MEDLINE | ID: mdl-29909078

ABSTRACT

Nitro-fatty acids (NFAs) are endogenously occurring lipid mediators exerting strong anti-inflammatory effects and acting as anti-oxidants in a number of animal models of inflammation. These NFA effects are mediated by targeting important regulatory proteins involved in inflammatory processes, such as 5-lipoxygenase, soluble epoxide hydrolase, or NF-κB. In the present study, we investigated the anti-tumorigenic effects of NFAs on colorectal cancer (CRC) cells in cell culture-based experiments and in a murine xenograft model of human CRC. We could show that 9-NOA suppresses the viability of CRC cells (HCT-116 and HT-29) by inducing a caspase-dependent apoptosis via the intrinsic apoptotic pathway. Co-treatment with the pan-caspase inhibitor Q-VD-OPH counteracted the NFA-mediated apoptosis in both cell lines. Furthermore, NFAs affected the cell cycle transition and reduced the oxygen consumption rate (OCR) immediately. On the contrary to their well-known anti-oxidative properties, NFAs mediated the generation of mitochondrial oxidative stress in human CRC cells. Additionally, similar to the cytostatic drug mitomycin, 9-NOA significantly reduced tumor growth in a murine xenograft model of human colorectal cancer. In contrast to the established cytostatic drug, 9-NOA treatment was well tolerated by mice. This study delivers a novel mechanistic approach for nitro-fatty acid-induced inhibition of CRC cell growth by targeting mitochondrial functions such as the mitochondrial membrane potential and mitochondrial respiration. We suggest these naturally occurring lipid mediators as a new class of well tolerated chemotherapeutic drug candidates for treatment of CRC or potentially other inflammation-driven cancer types.


Subject(s)
Anti-Inflammatory Agents/metabolism , Apoptosis/physiology , Cell Proliferation/physiology , Colorectal Neoplasms/metabolism , Fatty Acids/metabolism , Mitochondria/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Fatty Acids/pharmacology , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Male , Mice , Mice, Inbred ICR , Mitochondria/drug effects
9.
Front Pharmacol ; 9: 369, 2018.
Article in English | MEDLINE | ID: mdl-29755348

ABSTRACT

Early and adequate restoration of endothelial and tubular renal function is a substantial step during regeneration after ischemia reperfusion (IR) injury, occurring, e.g., in kidney transplantation, renal surgery, and sepsis. While tubular epithelial cell injury has long been of central importance, recent perception includes the renal vascular endothelium. In this regard, the fibrin cleavage product fibrinopeptide Bß15-42 mitigate IR injury by stabilizing interendothelial junctions through its affinity to VE-cadherin. Therefore, this study focused on the effect of Bß15-42 on post-acute physiological renal regeneration. For this, adult male C57BL/6 mice were exposed to a 30 min bilateral renal ischemia and reperfusion for 24 h or 48 h. Animals were randomized in a non-operative control group, two operative groups each treated with i.v. administration of either saline or Bß15-42 (2.4 mg/kg) immediately prior to reperfusion. Endothelial activation and inflammatory response was attenuated in renal tissue homogenates by single application of Bß15-42. Meanwhile, Bß15-42 did not affect acute kidney injury markers. Regarding the angiogenetic players VEGF-A, Angiopoietin-1, Angiopoietin-2, however, we observed significant higher expressions at mRNA and trend to higher protein level in Bß15-42 treated mice, compared to saline treated mice after 48 h of IR, thus pointing toward an increased angiogenetic activity. Similar dynamics were observed for the intermediate filament vimentin, the cytoprotective protein klotho, stathmin and the proliferation cellular nuclear antigen, which were significantly up-regulated at the same points in time. These results suggest a beneficial effect of anatomical contiguously located endothelial cells on tubular regeneration through stabilization of endothelial integrity. Therefore, it seems that Bß15-42 represents a novel pharmacological approach in the targeted therapy of acute renal failure in everyday clinical practice.

10.
Basic Clin Pharmacol Toxicol ; 122(2): 223-232, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28857508

ABSTRACT

Acute kidney injury remains an important cause of renal dysfunction. In this context, Toll-like receptors have been demonstrated to play a critical role in the induction of innate and inflammatory responses. Among these, Toll-like receptor 2 (TLR2) is constitutively expressed in tubular epithelial cells (TECs) of the kidney and is also known to mediate ischaemia reperfusion (IR) injury. Adult male C57BL/6JRj mice were randomized into seven groups (n = 8): a non-operative control group (CTRL) and six interventional groups in which mice were subjected to a 30 min. bilateral renal ischaemia. Immediately before reperfusion, mice were treated either with saline or with TLR2 antibody (clone T2.5) and harvested after ischaemia and reperfusion for 3, 24 and 48 hr. Analysed kidney homogenates of TLR2 antibody-treated mice displayed significantly decreased levels of TLR2 protein after 3 hr of IR compared to saline-treated mice. Accordingly, the degree of AKT phosphorylation was significantly decreased after 3 hr of IR compared to saline-treated animals. TUNEL staining revealed significantly higher apoptosis rates in TLR2 antibody-treated animals compared to saline-treated mice after 3 and 24 hr of IR. Further, a positive correlation between TLR2 protein expression and phosphorylation of AKT as well as a negative correlation with the number of TUNEL-positive cells could be observed. Inhibition of TLR2 and its signalling pathway by a single application of TLR2 antibody results in reduced phosphorylation of AKT and consecutively increased apoptosis.


Subject(s)
Acute Kidney Injury/drug therapy , Antibodies/pharmacology , Apoptosis/drug effects , Kidney/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/chemically induced , Toll-Like Receptor 2/antagonists & inhibitors , Acute Kidney Injury/enzymology , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Animals , Antibodies/toxicity , Disease Models, Animal , Kidney/enzymology , Kidney/immunology , Kidney/pathology , Lipocalin-2/genetics , Lipocalin-2/metabolism , Male , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Phosphorylation , Reperfusion Injury/enzymology , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Signal Transduction/drug effects , Time Factors , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism
11.
J Tissue Eng Regen Med ; 12(1): e486-e494, 2018 01.
Article in English | MEDLINE | ID: mdl-27689683

ABSTRACT

Shock wave therapy (SWT) has been shown to induce angiogenesis in ischaemic muscle. However, the mechanism of action remains unknown. Macrophages are crucial for angiogenic responses after ischaemic injury. The M2 macrophage subset enables tissue repair and induces angiogenesis. It was hypothesized that the angiogenic effects of SWT are at least partly caused by enhanced macrophage recruitment. C57BL/6 mice were subjected to hind limb ischaemia with subsequent SWT or sham treatment. Muscles were analysed via immunofluorescence staining, reverse-transcription polymerase chain reaction and western blot. Gene expression and proteins involved in macrophage recruitment were analysed and tissue sections were stained for macrophages, including subsets, capillaries and arterioles. Laser Doppler perfusion imaging was performed to assess functional outcome. Treated muscles showed increased expression of the pivotal macrophage recruiting factor monocyte chemotactic protein 1 (MCP-1). Higher levels of macrophage marker CD14 were found. Increased numbers of macrophages after SWT could be confirmed by immunofluorescence staining. The expression of the M2 polarization promoting chemokine interleukin 13 was significantly elevated in the treatment group. Elevated mRNA expression of the M2 scavenger receptor CD163 was found after SWT. Immunofluorescence staining confirmed increased numbers of M2 macrophages after treatment. It was found that SWT resulted in higher number of capillaries and arterioles. Assessment of functional outcome revealed significantly improved limb perfusion in treated animals. Shock wave therapy causes increased macrophage recruitment and enhanced polarization towards reparative M2 macrophages in ischaemic muscle resulting in angiogenesis and improved limb perfusion and therefore represents a promising new treatment option for the treatment of ischaemic heart disease. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
High-Energy Shock Waves , Hindlimb/blood supply , Ischemia/therapy , Macrophages/metabolism , Perfusion , Animals , Arterioles/pathology , Capillaries/pathology , Cell Count , Cell Polarity , Ischemia/pathology , Male , Mice, Inbred C57BL , Muscles/blood supply , Muscles/pathology
12.
Antioxid Redox Signal ; 28(14): 1265-1285, 2018 05 10.
Article in English | MEDLINE | ID: mdl-28699354

ABSTRACT

AIMS: 5-Lipoxygenase (5-LO) is the key enzyme of leukotriene (LT) biosynthesis and is critically involved in a number of inflammatory diseases such as arthritis, gout, bronchial asthma, atherosclerosis, and cancer. Because 5-LO contains critical nucleophilic amino acids, which are sensitive to electrophilic modifications, we determined the consequences of a drug-mediated intracellular release of nitric oxide (NO) on 5-LO product formation by human granulocytes and on 5-LO-dependent pulmonary inflammation in vivo. RESULTS: Clinically relevant concentrations of NO-releasing nonsteroidal anti-inflammatory drugs and other agents releasing NO intracellularly suppress 5-LO product synthesis in isolated human granulocytes via direct S-nitrosylation of 5-LO at the catalytically important cysteines 416 and 418. Furthermore, suppression of 5-LO product formation was observed in ionophore-stimulated human whole blood and in an animal model of pulmonary inflammation. INNOVATION: Here, we report for the first time that drugs releasing NO intracellularly are efficient 5-LO inhibitors in vitro and in vivo at least equivalent to approved 5-LO inhibitors. CONCLUSION: Our findings provide a novel mechanistic strategy for the development of a new class of drugs suppressing LT biosynthesis by site-directed nitrosylation. The results may also help to better understand the well-recognized anti-inflammatory clinically relevant actions of NO-releasing drugs. Furthermore, our study describes in detail a novel molecular mode of action of NO. Rebound Track: This work was rejected during standard peer review and rescued by Rebound Peer Review (Antioxid Redox Signal 16: 293-296, 2012) with the following serving as open reviewers: Angel Lanas, Hartmut Kühn, Joan Clària, Orina Belton. Antioxid. Redox Signal. 28, 1265-1285.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arachidonate 5-Lipoxygenase/metabolism , Leukotriene Antagonists/pharmacology , Leukotrienes/metabolism , Lipoxygenase Inhibitors/pharmacology , Nitric Oxide/pharmacology , Animals , Aspirin/pharmacology , Dose-Response Relationship, Drug , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Molecular Structure , Structure-Activity Relationship
13.
Transfusion ; 57(11): 2701-2711, 2017 11.
Article in English | MEDLINE | ID: mdl-28766731

ABSTRACT

BACKGROUND: The pathomechanisms of morbidity due to blood transfusions are not yet entirely understood. Elevated levels of red blood cell-derived microparticles (RMPs) are found in coagulation-related pathologies and also in stored blood. Previous research has shown that RMPs mediate transfusion-related complications by the intrinsic pathway. We hypothesized that RMPs might play a role in post-transfusion thrombotic complications by enhancing procoagulant activity also through the extrinsic pathway of coagulation. STUDY DESIGN AND METHODS: In this laboratory study, blood from 18 healthy volunteers was stimulated with microparticles from expired stored red blood cells. Various clotting parameters were recorded. Flow cytometry, enzyme-linked immunosorbent assays, and real-time polymerase chain reaction were used to investigate possible mediating mechanisms. RESULTS: The addition of RMPs shortened the clotting time from 194 to 161 seconds (p < 0.001). After incubation with RMPs, there was increased expression of tissue factor (TF) on monocytes and in plasma. TF messenger RNA expression increased in a time-dependent and concentration-dependent manner. There was a significant induction of interleukin-1ß and interleukin-6. After stimulation with RMPs, there was a significant increase in the number of activated platelets, an increased percentage of PAC-1/CD62P (procaspase activating compound-1/platelet surface P-selectin) double-positive platelets, and an increased number of platelet-neutrophil duplets and platelet-monocyte duplets, indicating enhanced interaction of platelets with neutrophils and monocytes. Levels of CXCL-8 (C-X-C motif chemokine ligand 1) and interleukin-6 were significantly higher after treatment with RMPs. CONCLUSION: Our results suggest that RMPs trigger coagulation through TF signaling, induce the secretion of proinflammatory cytokines, and induce cell-cell interaction between platelets and neutrophils. Thus, under certain conditions, RMPs could play a role in post-transfusion complications through these mechanisms.


Subject(s)
Blood Coagulation , Cell-Derived Microparticles/physiology , Erythrocytes/cytology , Inflammation , Transfusion Reaction , Blood Preservation , Cell Communication , Erythrocytes/ultrastructure , Humans , Inflammation/etiology , Interleukin-6/blood , Interleukin-8/blood , Monocytes/metabolism , Platelet Activation , RNA, Messenger/blood , Thromboplastin/genetics , Thromboplastin/metabolism , Thrombosis/etiology
14.
Int J Mol Sci ; 17(6)2016 May 24.
Article in English | MEDLINE | ID: mdl-27231899

ABSTRACT

Expression of kidney injury molecule-1 (Kim-1) is rapidly upregulated following tubular injury, constituting a biomarker for acute kidney damage. We examined the renal localization of Kim-1 expression in PKD/Mhm (polycystic kidney disease, Mannheim) (cy/+) rats (cy: mutated allel, +: wild type allel), an established model for autosomal dominant polycystic kidney disease, with chronic, mainly proximal tubulointerstitial alterations. For immunohistochemistry or Western blot analysis, kidneys of male adult heterozygously-affected (cy/+) and unaffected (+/+) littermates were perfusion-fixed or directly removed. Kim-1 expression was determined using peroxidase- or fluorescence-linked immunohistochemistry (alone or in combination with markers for tubule segments or differentiation). Compared to (+/+), only in (cy/+) kidneys, a chronic expression of Kim-1 could be detected by Western blot analysis, which was histologically confined to an apical cellular localization in areas of cystically-transformed proximal tubules with varying size and morphology, but not in distal tubular segments. Kim-1 was expressed by cystic epithelia exhibiting varying extents of dedifferentiation, as shown by double labeling with aquaporin-1, vimentin or osteopontin, yielding partial cellular coexpression. In this model, in contrast to other known molecules indicating renal injury and/or repair mechanisms, the chronic renal expression of Kim-1 is strictly confined to proximal cysts. Its exact role in interfering with tubulo-interstitial alterations in polycystic kidney disease warrants future investigations.


Subject(s)
Cell Adhesion Molecules/metabolism , Kidney Tubules, Proximal/metabolism , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Animals , Biomarkers/metabolism , Cell Dedifferentiation , Disease Models, Animal , Kidney Tubules, Proximal/pathology , Male , Organ Specificity , Rats , Up-Regulation
15.
Tumour Biol ; 37(7): 9649-56, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26797799

ABSTRACT

The oncogenic transcription factor signal transducer and activator of transcription 3 (STAT3) is a cytokine-activated transcription factor controlling inflammation, cell proliferation, survival, and differentiation in normal tissue as well as in tumor growth. One of its most important negative regulators is the suppressor of cytokine signaling 3 (SOCS3). Here, we analyzed SOCS3 and other tumor-associated local immune regulators in human clear cell renal cell carcinoma (ccRCC). Analyses were performed in tumor and adjacent tumor-free healthy renal tissue from 35 patients with ccRCC. For functional analysis, ccRCC Caki-1 cell lines were stimulated with IL-6 and IFNγ in cell culture assays. We observed significantly lower SOCS3 messenger RNA (mRNA) levels in tumor tissue compared to healthy tissue. SOCS3 mRNA strongly correlated within tumor and healthy tissue. Interestingly vice versa, SOCS3 protein levels were significantly higher in tumor tissue than in healthy tissue. IL-22 and IL-22R1 mRNA displayed no differences in tumor and healthy tissue. Stimulation of Caki-1 cells with IFNγ resulted in markedly increased SOCS3 mRNA levels. We conclude that SOCS3 along with STAT3 participates in regulatory mechanisms in ccRCC, which certainly features only one of multiple factors involved but nevertheless merits further attention.


Subject(s)
Carcinoma, Renal Cell/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Gene Expression Regulation/genetics , Humans , Inflammation/genetics , Interferon-gamma/genetics , Interleukin-6/genetics , Interleukins/genetics , Male , Middle Aged , RNA, Messenger/genetics , STAT3 Transcription Factor/genetics , Interleukin-22
16.
Cardiovasc Res ; 109(2): 331-43, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26676850

ABSTRACT

AIMS: Shock wave therapy (SWT) represents a clinically widely used angiogenic and thus regenerative approach for the treatment of ischaemic heart or limb disease. Despite promising results in preclinical and clinical trials, the exact mechanism of action remains unknown. Toll-like receptor 3, which is part of the innate immunity, is activated by binding double-stranded (ds) RNA. It plays a key role in inflammation, a process that is needed also for angiogenesis. We hypothesize that SWT causes cellular cavitation without damaging the target cells, thus liberating cytoplasmic RNA that in turn activates TLR3. METHODS AND RESULTS: SWT induces TLR3 and IFN-ß1 gene expression as well as RNA liberation from endothelial cells in a time-dependant manner. Conditioned medium from SWT-treated HUVECs induced TLR3 signalling in reporter cells. The response was lost when the medium was treated with RNase III to abolish dsRNAs or when TLR3 was silenced using siRNAs. In a mouse hind limb ischaemia model using wt and TLR3(-/-) mice (n = 6), SWT induced angiogenesis and arteriogenesis only in wt animals. These effects were accompanied by improved blood perfusion of treated limbs. Analysis of main molecules of the TLR3 pathways confirmed TLR3 signalling in vivo following SWT. CONCLUSION: Our data reveal a central role of the innate immune system, namely Toll-like receptor 3, to mediate angiogenesis upon release of cytoplasmic RNAs by mechanotransduction of SWT.


Subject(s)
Endothelial Cells/metabolism , Immunity, Innate/immunology , Inflammation/metabolism , Mechanotransduction, Cellular/physiology , Neovascularization, Pathologic/metabolism , Signal Transduction , Animals , Ischemia/metabolism , Male , Mice, Inbred C57BL , RNA, Double-Stranded/metabolism , Toll-Like Receptor 3/metabolism
17.
Pharmacol Ther ; 157: 43-64, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26549540

ABSTRACT

Canonical Wnt signaling is a highly conserved pathway with a prominent role in embryogenic development, adult tissue homeostasis, cell polarization, stem cell biology, cell differentiation, and proliferation. Furthermore, canonical Wnt signaling is of pivotal importance in the pathogenesis of a number of cancer types and crucially affects tumor initiation, cancer cell proliferation, cancer cell apoptosis, and metastasis. Reports over the last decade have provided strong evidence for a pathophysiological role of Wnt signaling in non-malignant classical inflammatory and neurodegenerative diseases. Although, several agents suppressing the Wnt pathway at different levels have been identified, the development of clinically relevant Wnt-inhibiting agents remains challenging due to selectivity and toxicity issues. Several studies have shown that long-term administration of non-steroidal anti-inflammatory drugs protects against colon cancer and potentially other tumor types by interfering both with the COX and the Wnt pathway. Our own studies have shown that non-steroidal anti-inflammatory drugs suppress Wnt signaling by targeting the pro-inflammatory enzyme 5-lipoxygenase which is the key enzyme pathophysiologically involved in the synthesis of leukotrienes. Furthermore, we found a direct link between the 5-lipoxygenase and Wnt signaling pathways, which is essential for the maintenance of leukemic stem cells. Accordingly, genetic and pharmacological inhibition of 5-lipoxygenase led to an impairment of Wnt-dependent acute and chronic myeloid leukemic stem cells. We believe that 5-lipoxygenase inhibitors might represent a novel type of Wnt inhibitor activating a potentially naturally occurring novel mechanism of suppression of Wnt signaling that is non-toxic, at least in mice, and is potentially well tolerated in patients.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Cyclooxygenase 2/metabolism , Neoplasms/metabolism , Wnt Proteins/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Inflammation/metabolism , Lipoxygenase Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway/drug effects
18.
Microvasc Res ; 101: 48-54, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26116861

ABSTRACT

The disruption of endothelial integrity is a crucial step for the development of vascular leakage and consequently ischemia-reperfusion injury (IRI). Regarding the molecular cell-cell interaction, the fibrinopeptide Bß15-42 prevents vascular leakage by stabilizing the inter-endothelial junctions via association with the vascular endothelial-cadherin. In a previous study we showed that a renoprotective effect in early IRI may be achieved by intravenous administration of Bß15-42 at the time of reperfusion. We now aimed to investigate whether additional pre-ischemic application of Bß15-42 could enhance this effect. Therefore C57BL/6 mice were subjected to 0.5h bilateral renal ischemia followed by reperfusion. The animals were randomized into 6 groups (n=6): two control groups treated with i.v. administration of NaCl at reperfusion for 0.5h (NaCl 1h) and 2.5h (NaCl 3h), two groups with Bß15-42 at reperfusion for 0.5h (Bß(rep) 1h) and 2.5h (Bß(rep) 3h), and two groups with administration of Bß15-42 immediately pre-ischemic as well as at reperfusion for 0.5h (Bß(peri) 1h) and 2.5h (Bß(peri) 3h). We found that both Bß(rep) and Bß(peri) mice displayed reduced early renal damage compared with NaCl treated mice. However, there was no further reduction of the IR damage through added pre-ischemic application of Bß15-42. Overall, we detected significantly reduced endothelial activation, lower tissue infiltration of neutrophils as well as lower tissue levels of neutrophil gelatinase-associated lipocalin (NGAL) in all mice treated with Bß15-42 compared to mice treated with NaCl. Our data confirm the renoprotective effect of Bß15-42 in the early therapeutic treatment of acute kidney injury due to ischemia and reperfusion. However, a combined pre-and post-ischemic administration of Bß15-42 appears to provide no additional benefit compared with a sole administration at reperfusion.


Subject(s)
Fibrin Fibrinogen Degradation Products/administration & dosage , Ischemia/drug therapy , Kidney/drug effects , Peptide Fragments/administration & dosage , Reperfusion Injury/drug therapy , Acute-Phase Proteins/metabolism , Animals , Capillary Permeability , Cell Adhesion , Endothelial Cells/metabolism , Immunohistochemistry , Inflammation/pathology , Kidney/pathology , Lipocalin-2 , Lipocalins/metabolism , Male , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Neutrophils/pathology , Oncogene Proteins/metabolism , Time Factors
19.
Int Urol Nephrol ; 46(12): 2243-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25218613

ABSTRACT

PURPOSE: We investigated whether the recently established biomarkers of acute kidney injury, neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1), may help to diagnose acute urinary tract infections (UTI) in adults and are able to distinguish between upper or lower localization. METHODS: NGAL levels were measured in blood and urine, and KIM-1 concentrations in urine of 97 subjects. We recruited age- and gender-matched groups of 30 patients with acute upper UTI and 29 patients with acute lower UTI as well as 38 healthy controls. NGAL and KIM-1 were determined by ELISA, serum and urine creatinine applying the Jaffé's method. RESULTS: Urinary NGAL (uNGAL) was significantly increased in patients with upper as well as with lower UTI compared with the healthy controls (P < 0.01; P < 0.05). Accordingly, uNGAL normalized on urinary creatinine (uNGAL/uCrea) was markedly higher in patients with lower UTI compared with the control group (P < 0.05), and uNGAL/uCrea levels were still raised in patients with upper UTI, though not reaching statistical significance (P = 0.07). However, neither uNGAL nor uNGAL/uCrea levels displayed significant differences between patients with upper or lower UTI (P = 0.75; P = 0.97). uKIM-1 levels were close to the detection limit and too low to reliably reveal differences between the three groups. CONCLUSIONS: While uKIM-1 seems not to serve as a helpful biomarker in this setting, increased levels of uNGAL indicate inflammatory processes in the urinary tract in adults. However, the determination of uNGAL levels does not allow to differentiate between upper and lower UTI.


Subject(s)
Acute-Phase Proteins/urine , Lipocalins/blood , Lipocalins/urine , Proto-Oncogene Proteins/blood , Proto-Oncogene Proteins/urine , Urinary Tract Infections/blood , Urinary Tract Infections/urine , Acute Disease , Adult , Biomarkers/blood , Biomarkers/urine , Creatinine/blood , Creatinine/urine , Diagnosis, Differential , Enzyme-Linked Immunosorbent Assay , Female , Hepatitis A Virus Cellular Receptor 1 , Humans , Lipocalin-2 , Male , Membrane Glycoproteins/urine , Receptors, Virus
20.
J Cancer Res Clin Oncol ; 140(10): 1671-80, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24903965

ABSTRACT

PURPOSE: While their negative impact on bone health is well established, the effects of aromatase inhibition (AI) on Wnt inhibitors and osteoprotegerin (OPG) are unknown. The aim of the study was to investigate the effects of AI on serum levels of sclerostin, DKK-1 and OPG, as well as their associations with PINP and CTX as markers of bone turnover and bone mineral density (BMD) assessed by DXA. METHODS: We conducted a prospective longitudinal analysis of 70 postmenopausal women with hormone receptor-positive early breast cancer (BC) treated with anastrozole. All measurements were performed at baseline, 12 and 24 months of treatment. We measured the association of the investigated variables with circulating bone turnover markers, as well as with the BMD. RESULTS: After 24 months of AI therapy, sclerostin and OPG concentrations increased from 29.5 pmol/l (SD = 15.1) and 6.8 pmol/l (SD = 2.2) at baseline to 43.2 pmol/l (SD = 20.6) (p < 0.001) and 7.4 pmol/l (SD = 2.2) (p = 0.028), respectively. DKK-1 levels decreased from 34.3 pmol/l (SD = 13.5) at baseline to 29.7 pmol/l (SD = 12.3) at the 24-month visit (p = 0.005). Sclerostin levels significantly correlated with PTH, OPG and BMD of the lumbar spine, while DKK-1 correlated with the BMD of the femoral neck and of the total hip. CONCLUSIONS: The observed increase in sclerostin levels indicates a central role of osteocytes in bone turnover in women with BC.


Subject(s)
Antineoplastic Agents/pharmacology , Aromatase Inhibitors/pharmacology , Biomarkers, Tumor/blood , Bone Density , Bone Morphogenetic Proteins/blood , Bone Remodeling , Breast Neoplasms/blood , Intercellular Signaling Peptides and Proteins/blood , Osteoprotegerin/blood , Adaptor Proteins, Signal Transducing , Aged , Bone Density/drug effects , Bone Neoplasms/blood , Bone Remodeling/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Genetic Markers , Humans , Middle Aged , Osteocytes/metabolism , Postmenopause , Prospective Studies , Time Factors , Wnt Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...