Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Urologie ; 62(2): 125-131, 2023 Feb.
Article in German | MEDLINE | ID: mdl-36690726

ABSTRACT

BACKGROUND: In many countries, such as France, England, USA, Canada, Australia, and New Zealand, alloplastic material in prolapse surgery has been paused due to the US Food and Drug Administration (FDA) warning, and restricted in other countries like the Netherlands and Sweden. For Europe and thus Germany, the SCENIHR report allows alloplastic material to be used for prolapse repair after recurrence and in other special situations. QUESTION: Which established and innovative prolapse surgeries without alloplastic material are currently available? METHODS: A literature search was carried out on established, guideline-compliant pelvic floor surgeries without alloplastic material as well as innovative new approaches. RESULTS: An established procedure for a defect in the anterior compartment is anterior colporrhaphy, which is associated with a high recurrence rate. The double-layered anterior colporrhaphy is a new approach and so far is associated with an improved 19-month outcome. Apical pelvic organ prolapse can be corrected by sacrouterine ligament fixation and vaginal sacrospinous fixation. New innovative techniques include laparoscopic unilateral pectineal suspension and the use of the semitendinosus tendon autograft to perform pectopexy or sacropexy. However, long-term data are still pending. In case of a posterior vaginal wall prolapse, posterior colporrhaphy is the therapy of choice and is associated with good success rates. CONCLUSION: Well-known surgical procedures with native tissue are experiencing a renaissance and new, innovative surgical approaches with good postoperative results are being developed. However, long-term studies are still necessary.


Subject(s)
Pelvic Organ Prolapse , Uterine Prolapse , United States , Female , Humans , Surgical Mesh , Pelvic Organ Prolapse/surgery , Uterine Prolapse/surgery , Vagina/surgery , Gynecologic Surgical Procedures/methods
2.
Scand J Immunol ; 74(2): 126-34, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21410503

ABSTRACT

γδ T cells play an important role in anti-infective immunity. The major subset of human γδ T cells selectively recognizes phosphorylated bacterial metabolites of the isoprenoid biosynthesis pathway, so-called phosphoantigens. The activation of γδ T cells is modulated by functionally expressed innate immune receptors, notably Toll-like receptor 2 and 3. It was also reported that in vitro expanded γδ T cells respond to muramyl dipeptide (MDP), the minimal peptidoglycan motif activating the nucleotide-binding oligomerization domain containing 2 (NOD2) receptor, although it is unknown whether ex vivo isolated human γδ T cells express functional NOD2. Here, we report that freshly isolated, highly purified peripheral blood γδ T cells express NOD2 mRNA and detectable amounts of NOD2 protein. The biologically active MDP L-D isomer but not the inactive D-D isomer augmented the interferon-γ (IFN-γ) secretion in phosphoantigen-stimulated peripheral blood mononuclear cells. Moreover, a moderate but reproducible and statistically significant increase in IFN-γ secretion was also observed when highly purified peripheral blood γδ T cells were activated by T cell receptor cross-linking in the presence of MDP. Taken together, our results indicate that in addition to the T cell receptor and Toll-like receptors, circulating human γδ T cells express NOD2 as a third class of pattern recognition receptor for sensing bacterial products.


Subject(s)
Nod2 Signaling Adaptor Protein/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Acetylmuramyl-Alanyl-Isoglutamine/immunology , Cells, Cultured , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Receptors, Pattern Recognition/immunology , T-Lymphocyte Subsets/metabolism
3.
Immunobiology ; 226(2): 152070, 2021 03.
Article in English | MEDLINE | ID: mdl-33639524

ABSTRACT

Signaling via TNF-R1 mediates pleiotropic biological outcomes ranging from inflammation and proliferation to cell death. Previous reports demonstrated that pro-survival signaling emanates from membrane resident TNF-R1 complexes (complex I) while only internalized TNF-R1 complexes are capable for DISC formation (complex II) and thus, apoptosis induction. Internalized TNF-R1 containing endosomes undergo intracellular maturation towards lysosomes, resulting in activation and release of Cathepsin D (CtsD) into the cytoplasm. We recently revealed HSP90 as target for proteolytic cleavage by CtsD, resulting in cell death amplification. In this study, we show that extrinsic cell death activation via TNF or TRAIL results in HSP90ß degradation. Co-incubation of cells with either TNF or TRAIL in combination with the HSP90ß inhibitor KUNB105 but not HSP90α selective inhibition promotes apoptosis induction. In an attempt to reveal further downstream targets of combined TNF-R1 or TRAIL-R1/-R2 activation with HSP90ß inhibition, we identify HIF1α and validate its ligand:inhibitor triggered degradation. Together, these findings suggest that selective inhibition of HSP90 isoforms together with death ligand stimulation may provide novel strategies for therapy of inflammatory diseases or cancer, in future.


Subject(s)
HSP90 Heat-Shock Proteins/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , Tumor Necrosis Factor-alpha/immunology , Apoptosis , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HeLa Cells , Humans , Proteome , U937 Cells
4.
J Exp Med ; 174(5): 975-88, 1991 Nov 01.
Article in English | MEDLINE | ID: mdl-1658188

ABSTRACT

Tumor necrosis factor (TNF) is a proinflammatory polypeptide that is able to induce a great diversity of cellular responses via modulating the expression of a number of different genes. One major pathway by which TNF receptors communicate signals from the membrane to the cell nucleus involves protein kinase C (PKC). In the present study, we have addressed the molecular mechanism of TNF-induced PKC activation. To this, membrane lipids of the human histiocytic cell line U937 were labeled by incubation with various radioactive precursors, and TNF-induced changes in phospholipid, neutral lipid, and water-soluble metabolites were analyzed by thin layer chromatography. TNF treatment of U937 cells resulted in a rapid and transient increase of 1'2'diacylglycerol (DAG), a well-known activator of PKC. The increase in DAG was detectable as early as 15 s after TNF treatment and peaked at 60 s. DAG increments were most pronounced (approximately 360% of basal levels) when cells were preincubated with [14C]lysophosphatidylcholine, which was predominantly incorporated into the phosphatidylcholine (PC) pool of the plasma-membranes. Further extensive examination of changes in metabolically labeled phospholipids indicated that TNF-stimulated hydrolysis of PC is accompanied by the generation of phosphorylcholine and DAG. These results suggest the operation of a PC-specific phospholipase C. Since no changes in phosphatidic acid (PA) and choline were observed and the production of DAG by TNF could not be blocked by either propranolol or ethanol, a combined activation of phospholipase D and PA-phosphohydrolase in DAG production appears unlikely. TNF-stimulated DAG production as well as PKC activation could be blocked by the phospholipase inhibitor p-bromophenacylbromide (BPB). Since BPB did not inactivate PKC directly, these findings underscore that TNF activates PKC via formation of DAG. TNF stimulation of DAG production could be inhibited by preincubation of cells with a monoclonal anti-TNF receptor (p55-60) antibody, indicating that activation of a PC-specific phospholipase C is a TNF receptor-mediated event.


Subject(s)
Diglycerides/biosynthesis , Phosphatidylcholines/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Type C Phospholipases/physiology , Calcium/metabolism , Enzyme Activation , Humans , Phospholipase D/analysis , Phospholipases A/physiology , Protein Kinase C/analysis , Receptors, Cell Surface/analysis , Receptors, Tumor Necrosis Factor
5.
J Exp Med ; 184(2): 725-33, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8760826

ABSTRACT

Tumor necrosis factor (TNF) is a pleiotropic mediator of inflammation that has been implicated in the pathogenesis of devastating clinical syndromes including septic shock. We have investigated the role of a TNF-responsive phosphatidylcholine-specific phospholipase C (PC-PLC) for the cytotoxic and proinflammatory activity of TNF. We show here that the cytotoxicity signaled for by the so-called "death domain" of the p55 TNF receptor is associated with the activation of PC-PLC. The xanthogenate tricyclodecan-9-yl (D609), a specific and selective inhibitor of PC-PLC, blocked the cytotoxic action of TNF on L929 and Wehi164 cells. In vivo, D609 prevented both adhesion molecule expression in the pulmonary vasculature and the accompanying leukocyte infiltration in TNF-treated mice. More strikingly, D609 protects BALB/c mice from lethal shock induced either by TNF, lipopolysaccharide, or staphylococcal enterotoxin B. Together these findings imply PC-PLC as an important mediator of the pathogenic action of TNF, suggesting that PC-PLC may serve as a novel target for anti-inflammatory TNF antagonists.


Subject(s)
Antigens, CD/physiology , Phosphatidylcholines/physiology , Receptors, Tumor Necrosis Factor/physiology , Type C Phospholipases/physiology , Animals , Bridged-Ring Compounds/pharmacology , Cell Line , Enterotoxins , Enzyme Activation , Mice , Norbornanes , Phosphodiesterase Inhibitors/pharmacology , Phospholipases A/metabolism , Receptors, Tumor Necrosis Factor, Type I , Shock, Septic/prevention & control , Signal Transduction , Thiocarbamates , Thiones/pharmacology , Type C Phospholipases/antagonists & inhibitors
6.
J Exp Med ; 181(6): 2059-68, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7759998

ABSTRACT

T cell receptor recognition of antigen can lead either to T lymphocyte differentiation and proliferation or to a state of unresponsiveness, which is dependent on whether appropriate costimulatory signals are provided to the mature T cell. We have investigated a novel intracellular signaling pathway provided by the costimulatory molecule CD28. CD28 engagement triggers the activation of an acidic sphingomyelinase (A-SMase), which results in the generation of ceramide, an important lipid messenger intermediate. A-SMase activation by CD28 occurred in resting as well as in activated primary T cells or leukemic Jurkat cells. In contrast, ligation of either CD3 or CD2 did not result in A-SMase activation. Overexpression of recombinant A-SMase in Jurkat T cells substituted for CD28 with regard to nuclear factor-kB activation. These data suggest that CD28 provides an important costimulatory signal by activation of an acidic sphingomyelinase pathway.


Subject(s)
CD28 Antigens/physiology , Receptors, Antigen, T-Cell/physiology , Signal Transduction , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocytes/physiology , Animals , Cell Differentiation , Cell Line , Cricetinae , Enzyme Activation , Female , Humans , Kinetics , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Phytohemagglutinins , T-Lymphocytes/cytology , T-Lymphocytes/enzymology
7.
J Clin Invest ; 104(12): 1761-70, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10606630

ABSTRACT

Epidermal TNF expression increases in response to cutaneous permeability barrier disruption and wound healing. TNF signaling is mediated by acid and neutral sphingomyelinases (A- and N-SMase), which generate ceramide, an important regulator of proliferation, differentiation, and apoptosis. In the epidermis, ceramide is known to be an integral part of the extracellular stratum corneum (SC) lipid bilayers that constitute the permeability barrier of the skin. We show here that topical application of TNF after experimental injury to the SC of hairless mice (hr(-/-)) enhances barrier repair. In TNF receptor p55-deficient (TNF-R55-deficient) mice (hr(+/+)), cutaneous barrier repair was delayed compared with wild-type (hr(+/+)) or TNF-R75-deficient (hr(+/+)) animals. After barrier disruption in hairless (hr(-/-)) and wild-type (hr(+/+)), but not in TNF-R55-deficient (hr(+/+)) mice, the enzymatic activities of both A-SMase and N-SMase were significantly enhanced. Stimulation of SMase activities was accompanied by an increase in C(24)-ceramide levels. Most A-SMase activity in hairless mice (hr(-/-)) was found in the outer epidermal cell layers and colocalized in the lamellar bodies with A-SMase and sphingomyelin. Reduction of epidermal A-SMase activity by the inhibitor imipramine resulted in delayed permeability barrier repair after SC injury. Together, these results suggest that TNF-R55 signaling pathways contribute to cutaneous permeability barrier repair through SMase-mediated generation of ceramide.


Subject(s)
Antigens, CD/physiology , Receptors, Tumor Necrosis Factor/physiology , Skin/metabolism , Sphingomyelin Phosphodiesterase/physiology , Animals , Ceramides/analysis , Glucosylceramidase/physiology , Imipramine/pharmacology , Male , Mice , Mice, Hairless , Mice, Inbred C57BL , Permeability , Receptors, Tumor Necrosis Factor, Type I , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelins/analysis , Tumor Necrosis Factor-alpha/pharmacology
8.
Cancer Res ; 48(19): 5417-21, 1988 Oct 01.
Article in English | MEDLINE | ID: mdl-3262009

ABSTRACT

In this paper we show that eight of 17 tumor cell lines of various tissue origin constitutively express tumor necrosis factor (TNF) mRNA. Five of these eight cell lines concomitantly contained lymphotoxin (LT) mRNA. Of the remaining nine cell lines that lacked detectable TNF or LT gene expression, five could be induced by phorbol ester and/or cytokines to accumulate the respective mRNAs. While TNF mRNA was found not only in neoplastic hematopoietic cells, but also in cell lines derived from carcinomas, LT gene expression seemed to be restricted to lymphoid tumor cells. Tumor cells that expressed LT mRNA also secreted LT protein and proved to be resistant to the cytostatic/cytotoxic effects of their own protein product as well as to exogenous recombinant TNF and recombinant LT. In contrast, most of the cell lines containing TNF mRNA did not release TNF protein into the supernatants, indicating that TNF gene expression may be controlled predominantly at a post-transcriptional level. Thus, the presence of TNF mRNA does not necessarily reflect a TNF-resistant phenotype. Our findings demonstrate that TNF and/or LT mRNA expression is a rather common phenomenon in long-term cultured tumor cell lines and reveal that ectopic TNF and/or LT production by tumor cells may be involved in certain paraneoplastic syndromes as well as in tumorigenesis.


Subject(s)
Gene Expression Regulation , Lymphotoxin-alpha/genetics , Tumor Cells, Cultured/drug effects , Tumor Necrosis Factor-alpha/genetics , Cell Line , Humans , Phenotype , Tumor Cells, Cultured/metabolism
9.
Andrology ; 4(3): 430-41, 2016 05.
Article in English | MEDLINE | ID: mdl-26916383

ABSTRACT

Sperm chromatin structure and condensation determine accessibility for damage, and hence success of fertilization and development. The aim of this study was to reveal characteristic spectral features coinciding with abnormal sperm chromatin packing (i.e., DNA-protein interactions) and decreased fertility, using Fourier transform infrared spectroscopy. Chromatin structure in spermatozoa obtained from different stallions was investigated. Furthermore, spermatozoa were exposed to oxidative stress, or treated with thiol-oxidizing and disulfide-reducing agents, to alter chromatin structure and packing. Spectroscopic studies were corroborated with flow cytometric analyses using the DNA-intercalating fluorescent dye acridine orange. Decreased fertility of individuals correlated with increased abnormal sperm morphology and decreased stability toward induced DNA damage. Treatment with the disulfide reducing agent dithiothreitol resulted in increased sperm chromatin decondensation and DNA accessibility, similar as found for less mature epididymal spermatozoa. In situ infrared spectroscopic analysis revealed that characteristic bands arising from the DNA backbone (ν1230, ν1086, ν1051 cm(-1) ) changed in response to induced oxidative damage, water removal, and decondensation. This coincided with changes in the amide-I region (intensity at ν1620 vs. ν1640 cm(-1) ) denoting concomitant changes in protein secondary structure. Reduction in protein disulfide bonds resulted in a decreased value of the asymmetric to symmetric phosphate band intensity (ν1230/ν1086 cm(-1) ), suggesting that this band ratio is sensitive for the degree of chromatin condensation. Moreover, when analyzing spermatozoa from different individuals, it was found that the asymmetric/symmetric phosphate band ratio negatively correlated with the percentage of morphologically abnormal spermatozoa.


Subject(s)
Chromatin/chemistry , DNA Damage , Fertility/physiology , Spectroscopy, Fourier Transform Infrared , Spermatozoa/chemistry , Animals , Horses , Male , Oxidative Stress/physiology , Sperm Motility/physiology
10.
Oncogene ; 20(31): 4258-69, 2001 Jul 12.
Article in English | MEDLINE | ID: mdl-11464292

ABSTRACT

The molecular alterations in tumour cells leading to resistance towards apoptosis induced by CD95 and TRAIL-receptors are not fully understood. We report here that the stimulation of the CD95- and TRAIL-resistant human pancreatic adenocarcinoma cell line PancTuI with an agonistic anti-CD95 antibody or TRAIL resulted in activation of protein kinase C and NF-kappaB. Inhibition of protein kinase C by Gö6983 sensitized these cells to apoptotic challenges and strongly diminished activation of NF-kappaB by anti-CD95 and TRAIL. Similarly, inhibition of NF-kappaB by MG132 or by transient transfection with a dominant negative mutant of IkappaBalpha restored the responsiveness of PancTuI cells to both death ligands. In the CD95 and TRAIL-sensitive cell line Colo357 the induction of protein kinase C and NF-kappaB following activation of CD95 and TRAIL-R was very moderate compared with PancTuI cells. However, pre-incubation of these cells with PMA strongly reduced their apoptotic response to anti-CD95 and TRAIL. Taken together, we show that activation of protein kinase C operates directly in a death receptor-dependent manner in PancTuI cells and protect pancreatic tumour cells from anti-CD95 and TRAIL-mediated apoptosis by preventing the loss DeltaPsim and Cytochrome c release as well as by induction of NF-kappaB.


Subject(s)
Adenocarcinoma/pathology , Apoptosis/physiology , Membrane Glycoproteins/physiology , NF-kappa B/metabolism , Pancreatic Neoplasms/pathology , Protein Kinase C/metabolism , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology , Adenocarcinoma/enzymology , Adenocarcinoma/metabolism , Apoptosis Regulatory Proteins , Enzyme Activation , Humans , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured
11.
Cell Death Differ ; 11(5): 550-63, 2004 May.
Article in English | MEDLINE | ID: mdl-14739942

ABSTRACT

Acidic noncaspase proteases-like cathepsins have been introduced as novel mediators of apoptosis. A clear role for these proteases and the acidic endolysosomal compartment in apoptotic signalling is not yet defined. To understand the role and significance of noncaspases in promoting and mediating cell death, it is important to determine whether an intersection of these proteases and the caspase pathway exists. We recently identified the endolysosomal aspartate protease cathepsin D (CTSD) as a target for the proapoptotic lipid ceramide. Here, we show that tumor necrosis factor (TNF)-induced CTSD activation depends on functional acid sphingomyelinase (A-SMase) expression. Ectopic expression of CTSD in CTSD-deficient fibroblasts results in an enhanced TNF-mediated apoptotic response. Intracellular colocalization of CTSD with the proapoptotic bcl-2 protein family member Bid in HeLa cells, and the ability of CTSD to cleave directly Bid in vitro as well as the lack of Bid activation in cathepsin-deficient fibroblasts indicate that Bid represents a direct downstream target of CTSD. Costaining of CTSD and Bid with Rab5 suggests that the endosomal compartments are the common 'meeting point'. Caspase-9 and -3 activation also was in part dependent on A-SMase and CTSD expression as revealed in the respective deficiency models. Our results link as novel endosomal intermediates the A-SMase and the acid aspartate protease CTSD to the mitochondrial apoptotic TNF pathway.


Subject(s)
Carrier Proteins/metabolism , Caspases/metabolism , Cathepsin D/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein , Caspase 3 , Caspase 9 , Cells, Cultured , Ceramides/metabolism , Enzyme Activation/physiology , Female , Fibroblasts/metabolism , HeLa Cells , Humans , Mice , Mice, Knockout , Mitochondria/metabolism , Sphingomyelin Phosphodiesterase/metabolism
12.
J Leukoc Biol ; 56(5): 533-41, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7964160

ABSTRACT

Tumor necrosis factor (TNF) and interleukin-1 (IL-1) are cytokines with pleiotropic biological activities, exerting a broad range of overlapping biological functions. The redundancy of TNF and IL-1 activities may be based on the utilization of shared key components of intracellular signaling pathways. Two lipid second messengers have been found to transmit TNF and IL-1 intracellular signals: 1,2-diacylglycerol (DAG), generated by a phosphatidylcholine-specific phospholipase C, and ceramide, generated by sphingomyelinase (SMase). DAG is a well established activator of the important signaling system protein kinase C (PKC), which appears to mediate various cellular responses to TNF or IL-1. In addition, it is obvious that DAG also activates other enzyme systems like acidic sphingomyelinase. SMases have been implicated in a number of TNF responses, including stimulation of cell growth and differentiation, as well as triggering cytotoxicity and apoptosis. The metabolic active cleavage product of SMase, ceramide, is a novel multifunctional lipid second messenger capable of inducing various signaling systems. Both cytokines, TNF and IL-1, stimulate a neutral,plasma membrane-associated SMase that leads to stimulation of a protein kinase and eventually to activation of the mitogen-activated protein (MAP) kinase cascade and phospholipase A2. Ceramide is also capable of stimulating a cytosolic protein phosphatase. PKC plays a role in activation of the nuclear transcription factor AP-1, and the DAG-regulated acidic SMase is involved in transducing TNF signals to the cell nucleus via activation of the nuclear transcription factor NF-kappa B.


Subject(s)
Ceramides/biosynthesis , Diglycerides/biosynthesis , Interleukin-1/physiology , Protein Kinase C/physiology , Second Messenger Systems/physiology , Sphingomyelin Phosphodiesterase/metabolism , Tumor Necrosis Factor-alpha/physiology , Type C Phospholipases/metabolism , Animals , Enzyme Activation , Humans , NF-kappa B/metabolism
13.
J Invest Dermatol ; 115(4): 708-13, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10998148

ABSTRACT

Point mutations in the suprabasal cytokeratins 1 (K1) or 10 (K10) in humans have been shown to be the cause of the congenital ichthyosis epidermolytic hyperkeratosis. Recently, a K10 deficient mouse model was established serving as a model for epidermolytic hyperkeratosis. Homozygotes suffered from severe skin fragility and died shortly after birth. Heterozygotes developed hyperkeratosis with age. To see whether phenotypic abnormalities in the mouse model were associated with changes in skin barrier function and skin water content we studied basal transepidermal water loss and capacity for barrier repair after experimental barrier disruption as well as stratum corneum hydration. Also, we determined the activities of acid and neutral sphingomyelinase key enzymes of the tumor necrosis factor and interleukin-1 signal transduction pathways generating the ceramides most important for epidermal permeability barrier homeostasis. Neonatal homozygotes showed an 8-fold increase in basal transepidermal water loss compared with wild type controls. Adult heterozygotes exhibited delayed barrier repair after experimental barrier disruption. Stratum corneum hydration was reduced in homozygous and heterozygous mice. Acid sphingomyelinase activity, which is localized in the epidermal lamellar bodies and generates ceramides for extracellular lipid lamellae in the stratum corneum permeability barrier, was reduced in homozygous as well as heterozygous animals. Neutral sphingomyelinase activity, which has a different location and generates ceramides involved in cell signaling, was increased. The reduction in acid sphingomyelinase activity may explain the recently described decreased ratio of ceramides to total lipids in K10 deficient mice. In summary, our results demonstrate the crucial role of the keratin filament for permeability barrier function and stratum corneum hydration.


Subject(s)
Body Water/metabolism , Cell Membrane Permeability/physiology , Keratins/deficiency , Skin/cytology , Sphingomyelin Phosphodiesterase/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Heterozygote , Homeostasis , Homozygote , Humans , Hyperkeratosis, Epidermolytic , Mice , Skin/enzymology , Skin/metabolism
14.
FEBS Lett ; 443(1): 75-9, 1999 Jan 22.
Article in English | MEDLINE | ID: mdl-9928956

ABSTRACT

The angiotensin AT2 receptor has been implicated in both regeneration and apoptosis. To further investigate the molecular mechanisms leading to AT2 receptor-induced programmed cell death in PC12W cells we studied the effects of angiotensin II (ANG II) on ceramide levels by HPTLC analysis. We could demonstrate that ANG II time- (1-10 h) and dose-dependently (10(-8)-5 X 10(-6) M) increased ceramide levels by maximally 175% but did not affect sphingomyelin degradation. The ANG 11 effects were mediated by AT2 receptors since they were completely abolished by co-incubation with the AT2 receptor antagonist, PD123177 (10(-5) M), but not by the AT1 receptor antagonist, losartan (10(-5) M). These data suggest a novel signal transduction pathway to the AT2 receptor leading to apoptosis in neuronal cells.


Subject(s)
Angiotensin II/pharmacology , Apoptosis , Ceramides/metabolism , Receptors, Angiotensin/metabolism , Angiotensin II/antagonists & inhibitors , Angiotensin II Type 1 Receptor Blockers , Angiotensin II Type 2 Receptor Blockers , Angiotensin Receptor Antagonists , Animals , Chromaffin Cells , Dose-Response Relationship, Drug , Imidazoles/pharmacology , PC12 Cells , Pyridines/pharmacology , Rats , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Signal Transduction , Sphingomyelins/metabolism
15.
Semin Oncol ; 19(2 Suppl 4): 16-24, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1313193

ABSTRACT

Tumor necrosis factor (TNF) is able to induce a great diversity of cellular responses via modulating the expression of a number of different genes. The multitude of TNF activities may be explained by both structural and functional heterogeneity in TNF receptors as well as by a diversification of postreceptor signal transduction pathways. Purification of TNF receptors has revealed two major, distinct binding proteins (TR60 and TR80). TR60 seems to be an essential component for TNF signaling; the functional role of TR80 remains to be elucidated. The pathway of postreceptor signal transduction involves phospholipase A2, a phosphatidylcholine-specific phospholipase C, protein kinase C, and other serine/threonine and tyrosine-specific protein kinases with as yet unknown function. At the receiving end of TNF signaling, induction of gene expression is mediated through activation of nuclear transcription factors, such as NFkB, AP-1, IRF-1, and NF-GMa.


Subject(s)
Protein Kinase C/biosynthesis , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , Diglycerides/biosynthesis , Enzyme Activation , Phospholipases A/biosynthesis , Phospholipases A2 , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/physiology , Receptors, Tumor Necrosis Factor , Substrate Specificity , Transcription Factors/biosynthesis , Type C Phospholipases/biosynthesis
16.
Immunobiology ; 193(2-4): 193-203, 1995 Jul.
Article in English | MEDLINE | ID: mdl-8530143

ABSTRACT

Tumor Necrosis Factor (TNF) is one of the most potent physiological inducers of the nuclear transcription factor NF-kappa B. In light of the pivotal role of NF-kappa B in the development of immune responses and activation of HIV replication, the identification of TNF signal transduction pathways involved in NF-kappa B activation is of particular interest. Data from our laboratory demonstrate that the TNF signal transduction pathway-mediating NF-kappa B activation involves two phospholipases, a phosphatidylcholine-specific phospholipase C (PC-PLC) and an endosomal acidic sphingomyelinase (aSMase). The aSMase activation by TNF is secondary to the generation of 1,2-diacylglycerol (DAG) produced by a TNF-responsive PC-PLC. SMase and its product ceramide induce degradation of the NF-kappa B inhibitor I kappa B as well as NF-kappa B activation. Besides endosomal acidic SMase, TNF also rapidly activates a plasmamembrane-associated neural SMase (nSMase), that, however is not involved in TNF-induced NF-kappa B activation. NSMase and aSMase are activated by different cytoplasmic domains of the 55 kDa TNF-receptor and are coupled to select pathways of TNF signaling. Ceramide generated by nSMase directs the activation of proline-directed serin/threonine protein kinases and phospholipase A2 and ceramide produced by aSMase triggers the activation of NF-kappa B. No apparent crosstalk was detected between nSMase and aSMase pathways, indicating that ceramide action depends on the topology of its production.


Subject(s)
NF-kappa B/biosynthesis , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/physiology , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , NF-kappa B/drug effects , Signal Transduction/drug effects
17.
Pediatr Infect Dis J ; 16(5): 482-5, 1997 May.
Article in English | MEDLINE | ID: mdl-9154541

ABSTRACT

BACKGROUND: An increase in the number of patients presenting with extraintestinal salmonellosis has occurred at our institution. The purpose of this study was to review the extraintestinal salmonellosis cases in our institution and to investigate the possible reasons for this increase. METHODS: A retrospective review of patients from 1985 through 1996 was carried out to identify patients with extraintestinal infections with Salmonella. Demographic data were gathered and statistical evaluations comparing differences among groups (1985 to 1989, 1989 to 1992, 1993 to 1996) was done using the G statistic, adjusted (maximum likelihood) chi square or the Fisher's exact test. RESULTS: Thirty-nine patients were identified with extraintestinal salmonellosis and although the number of cases had increased from 8 in 1985 to 1988 to 18 in 1989 to 1992 and 13 from 1993 to 1996, the percentage of cases that were extraintestinal were similar (3.3%, 6%, 4.9%; P > 0.1). Seventy-two percent of patients had underlying risk factors with the most common being age < 3 months (44%), sickle cell anemia (13%) and gastrointestinal surgery (10%). Fever and diarrhea were more common presenting symptoms in patients < 3 months of age than in older patients (P < 0.05). Salmonella typhimurium and Salmonella heidelberg were the most common serotypes isolated and an increasing trend of ampicillin resistance was noted from 0% in 1985 to 1988 to 39% from 1989 to 1992 and 23% from 1993 to 1996. CONCLUSIONS: The reasons for an increasing trend in extraintestinal cases of human salmonellosis at our institution were not identified. This illness continues to occur in infants and children with well-recognized risk factors.


Subject(s)
Cross Infection/epidemiology , Hospitals, Pediatric/statistics & numerical data , Salmonella Infections/epidemiology , Adolescent , Adult , Age Distribution , Chi-Square Distribution , Confidence Intervals , Cross Infection/diagnosis , Cross Infection/drug therapy , Cross Infection/microbiology , Drug Resistance, Microbial , Female , Humans , Incidence , Infant , Infant, Newborn , Male , Retrospective Studies , Risk Factors , Salmonella/classification , Salmonella Infections/diagnosis , Salmonella Infections/drug therapy , Salmonella Infections/microbiology , Serotyping , Sex Distribution , United States/epidemiology
18.
Regul Pept ; 102(2-3): 93-9, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11730981

ABSTRACT

Nerve growth factor (NGF) has been previously shown to induce exocytosis in rat peritoneal mast cells (RPMCs) in the presence of lyso-phosphatidylserine (lysoPS) by interacting with high-affinity NGF receptors of the TrkA-type. In RPMCs, type D phosphatidylcholine-selective phospholipases (PLDs) have been postulated to be involved in some exocytotic signaling pathways induced by different agonists. The aim of the present study was to assess a putative functional role of PLD for NGF/lysoPS-induced exocytosis in RPMCs. In 1-[14C]palmitoyl-2-lyso-3-phosphatidylcholine-labelled RPMCs, NGF/lysoPS stimulated the formation of diacylglycerol (DAG) and, in the presence of ethanol (1% [v/v]), phosphatidylethanol (PEtOH). These data indicate PLD-activation by NGF/lysoPS in RPMCs. Preincubation of RPMCs for 2 min with ethanol, an inhibitor of PLD-derived DAG-formation, dose-dependently (IC(50): 0.6% [v/v]) and agonist-selectively inhibited the NGF/lysoPS induced release of [3H]serotonin ([3H]5-HT) in [3H]5-HT-loaded RPMCs, confirming the functional importance of PLD-action. Exocytosis and PEtOH-production was potently inhibited by the broad-spectrum serine/threonine kinase inhibitor staurosporine and activated by the protein kinase C(PKC)-activator PMA (phorbol-12-myristate-13-acetate) suggesting a role for PKC as mediator for NGF/lysoPS-induced activation of PLD.


Subject(s)
Exocytosis/drug effects , Lysophospholipids/pharmacology , Mast Cells/drug effects , Nerve Growth Factor/pharmacology , Phospholipase D/metabolism , Signal Transduction/drug effects , Animals , Diglycerides/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/pharmacology , Ethanol/pharmacology , Intercellular Signaling Peptides and Proteins , Mast Cells/cytology , Mast Cells/enzymology , Peptides , Peritoneum/cytology , Peritoneum/drug effects , Phorbol Esters/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Serotonin/metabolism , Time Factors , Wasp Venoms/pharmacology
19.
Adv Exp Med Biol ; 477: 305-15, 2000.
Article in English | MEDLINE | ID: mdl-10849758

ABSTRACT

We have identified the aspartic protease cathepsin D as a novel intracellular target protein for the lipid second messenger ceramide. Ceramide specifically binds to and induces CTSD proteolytic activity. A-SMase deficient cells derived from Niemann-Pick patients show decreased CTSD activity that was reconstituted by transfection with A-SMase cDNA. Ceramide accumulation in cells derived from A-ceramidase defective Farber patients correlates with enhanced CTSD activity. These findings suggest that A-SMase-derived ceramide targets endolysosomal CTSD.


Subject(s)
Cathepsin D/metabolism , Ceramides/physiology , Second Messenger Systems/physiology , Sphingomyelin Phosphodiesterase/physiology , Amidohydrolases/deficiency , Amidohydrolases/genetics , Animals , Apoptosis , Cathepsin D/deficiency , Cathepsin D/genetics , Cell Compartmentation , Cell Line, Transformed , Ceramidases , Ceramides/pharmacology , Cytokines/physiology , DNA, Complementary/genetics , Enzyme Activation/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , HeLa Cells/metabolism , Humans , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/metabolism , Lipid Metabolism , Lymphocytes/enzymology , Lysosomes/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Niemann-Pick Diseases/enzymology , Niemann-Pick Diseases/genetics , Niemann-Pick Diseases/pathology , Recombinant Fusion Proteins/metabolism , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Tumor Cells, Cultured , U937 Cells/metabolism
20.
Physiol Res ; 62(1): 95-105, 2013.
Article in English | MEDLINE | ID: mdl-23173681

ABSTRACT

Heterologous expression of Kir channels offers a tool to modulate excitability of neurons which provide insight into Kir channel functions in general. Inwardly-rectifying K+ channels (Kir channels) are potential candidate proteins to hyperpolarize neuronal cell membranes. However, heterologous expression of inwardly-rectifying K+ channels has previously proven to be difficult. This was mainly due to a high toxicity of the respective Kir channel expression. We investigated the putative role of a predominantly glial-expressed, weakly rectifying Kir channel (Kir4.1 channel subunit; KCNJ10) in modulating electrophysiological properties of a motoneuron-like cell culture (NSC-34). Transfection procedures using an EGFP-tagged Kir4.1 protein in this study proved to have no toxic effects on NSC-34 cells. Using whole cell-voltage clamp, a substantial increase of inward rectifying K+ currents as well as hyperpolarization of the cell membrane was observed in Kir4.1-transfected cells. Na+ inward currents, observed in NSC-34 controls, were absent in Kir4.1/EGFP motoneuronal cells. The Kir4.1-transfection did not influence the NaV1.6 sodium channel expression. This study demonstrates the general feasibility of a heterologous expression of a weakly inward-rectifying K+ channel (Kir4.1 subunit) and shows that in vitro overexpression of Kir4.1 shifts electrophysiological properties of neuronal cells to a more glial-like phenotype and may therefore be a candidate tool to dampen excitability of neurons in experimental paradigms.


Subject(s)
Neuroblastoma/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Spinal Cord Neoplasms/metabolism , Animals , Cell Line, Tumor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Membrane Potentials , Mice , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Neuroblastoma/genetics , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics , Recombinant Fusion Proteins/metabolism , Spinal Cord Neoplasms/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL