Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(23)2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38069220

ABSTRACT

Phosphatidylinositol-5-phosphate 4-kinase type 2 (PIP4K2) protein family members (PIP4K2A, PIP4K2B, and PIP4K2C) participate in the generation of PIP4,5P2, which acts as a secondary messenger in signal transduction, a substrate for metabolic processes, and has structural functions. In patients with acute myeloid leukemia (AML), high PIP4K2A and PIP4K2C levels are independent markers of a worse prognosis. Recently, our research group reported that THZ-P1-2 (PIP4K2 pan-inhibitor) exhibits anti-leukemic activity by disrupting mitochondrial homeostasis and autophagy in AML models. In the present study, we characterized the expression of PIP4K2 in the myeloid compartment of hematopoietic cells, as well as in AML cell lines and clinical samples with different genetic abnormalities. In ex vivo assays, PIP4K2 expression levels were related to sensitivity and resistance to several antileukemia drugs and highlighted the association between high PIP4K2A levels and resistance to venetoclax. The combination of THZ-P1-2 and venetoclax showed potentiating effects in reducing viability and inducing apoptosis in AML cells. A combined treatment differentially modulated multiple genes, including TAp73, BCL2, MCL1, and BCL2A1. In summary, our study identified the correlation between the expression of PIP4K2 and the response to antineoplastic agents in ex vivo assays in AML and exposed vulnerabilities that may be exploited in combined therapies, which could result in better therapeutic responses.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Humans , Cell Line, Tumor , Proto-Oncogene Proteins c-bcl-2/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Apoptosis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Phosphotransferases (Alcohol Group Acceptor)/pharmacology
2.
Immun Inflamm Dis ; 11(9): e998, 2023 09.
Article in English | MEDLINE | ID: mdl-37773702

ABSTRACT

AIM: Endoplasmic reticulum (ER) stress is common in different human pathologies, including cardiac diseases. Sphingosine kinase-1 (SPHK1) represents an important player in cardiac growth and function. Nevertheless, its function in cardiomyocyte ER stress remains vague. This study sought to evaluate the mechanism through which SPHK1 might influence ER stress during myocardial infarction (MI). METHODS: MI-related GEO data sets were queried to screen differentially expressed genes. Murine HL-1 cells exposed to oxygen-glucose deprivation (OGD) and mice with MI were induced, followed by gene expression manipulation using short hairpin RNAs and overexpression vectors. The activating transcription factor 3 (ATF3) and SPHK1 expression was examined in cells and tissues. Cell counting kit-8, TUNEL, DHE, HE, and Masson's staining were conducted in vitro and in vivo. The inflammatory factor concentrations in mouse serum were measured using ELISA. Finally, the transcriptional regulation of SPHK1 by ATF3 was validated. RESULTS: ATF3 and SPHK1 were upregulated in vivo and in vitro. ATF3 downregulation reduced the SPHK1 transcription. ATF3 and SPHK1 downregulation increased the viability of OGD-treated HL-1 cells and decreased apoptosis, oxidative stress, and ER stress. ATF3 and SPHK1 downregulation narrowed the infarction area and attenuated myocardial fibrosis in mice, along with reduced inflammation in the serum and ER stress in the myocardium. In contrast, SPHK1 reduced the protective effect of ATF3 downregulation in vitro and in vivo. CONCLUSIONS: ATF3 downregulation reduced SPHK1 expression to attenuate cardiomyocyte injury in MI.


Subject(s)
Activating Transcription Factor 3 , Myocytes, Cardiac , Mice , Humans , Animals , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 3/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Endoplasmic Reticulum Stress
3.
Int J Mol Sci ; 22(8)2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33917258

ABSTRACT

Cataracts are the major cause of blindness worldwide, largely resulting from aging and diabetes mellitus. Advanced glycation end products (AGEs) have been identified as major contributors in cataract formation because they alter lens protein structure and stability and induce covalent cross-linking, aggregation, and insolubilization of lens crystallins. We investigated the potential of the deglycating enzyme fructosamine-3-kinase (FN3K) in the disruption of AGEs in cataractous lenses. Macroscopic changes of equine lenses were evaluated after ex vivo intravitreal FN3K injection. The mechanical properties of an equine lens pair were evaluated after treatment with saline and FN3K. AGE-type autofluorescence (AF) was measured to assess the time-dependent effects of FN3K on glycolaldehyde-induced AGE-modified porcine lens fragments and to evaluate its actions on intact lenses after in vivo intravitreal FN3K injection of murine eyes. A potential immune response after injection was evaluated by analysis of IL-2, TNFα, and IFNγ using an ELISA kit. Dose- and time-dependent AF kinetics were analyzed on pooled human lens fragments. Furthermore, AF measurements and a time-lapse of macroscopic changes were performed on intact cataractous human eye lenses after incubation with an FN3K solution. At last, AF measurements were performed on cataractous human eyes after crossover topical treatment with either saline- or FN3K-containing drops. While the lenses of the equine FN3K-treated eyes appeared to be clear, the saline-treated lenses had a yellowish-brown color. Following FN3K treatment, color restoration could be observed within 30 min. The extension rate of the equine FN3K-treated lens was more than twice the extension rate of the saline-treated lens. FN3K treatment induced significant time-dependent decreases in AGE-related AF values in the AGE-modified porcine lens fragments. Furthermore, in vivo intravitreal FN3K injection of murine eyes significantly reduced AF values of the lenses. Treatment did not provoke a systemic immune response in mice. AF kinetics of FN3K-treated cataractous human lens suspensions revealed dose- and time-dependent decreases. Incubation of cataractous human eye lenses with FN3K resulted in a macroscopic lighter color of the cortex and a decrease in AF values. At last, crossover topical treatment of intact human eyes revealed a decrease in AF values during FN3K treatment, while showing no notable changes with saline. Our study suggests, for the first time, a potential additional role of FN3K as an alternative treatment for AGE-related cataracts.


Subject(s)
Cataract/drug therapy , Cataract/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Animals , Cataract/diagnosis , Cataract/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation , Eye/drug effects , Eye/metabolism , Glycation End Products, Advanced/administration & dosage , Horses , Humans , Immunohistochemistry , Intravitreal Injections , Lens, Crystalline/drug effects , Lens, Crystalline/metabolism , Mice , Phosphotransferases (Alcohol Group Acceptor)/administration & dosage , Phosphotransferases (Alcohol Group Acceptor)/therapeutic use
4.
Neuropharmacology ; 181: 108326, 2020 12 15.
Article in English | MEDLINE | ID: mdl-32966847

ABSTRACT

We have previously shown that sphingosine kinase 2 (SPK2) interacts with Bcl-2 via its BH3 domain, activating autophagy by inducing the dissociation of Beclin-1/Bcl-2 complexes, and that a TAT-SPK2 peptide containing the BH3 domain of SPK2 protects neurons against ischemic injury. The goals of the present study were to establish the functional significance of these findings, by testing whether TAT-SPK2 was effective in a mouse model of ischemic stroke, and to explore potential underlying mechanisms. Mice were administered with TAT-SPK2 by intraperitoneal injection before or after transient middle cerebral artery occlusion (tMCAO). Infarct volume, neurological deficit and brain water content were assessed 24 h after reperfusion. Mitophagy inhibitor Mdivi-1 and BNIP3 siRNAs were used to examine the involvement of BNIP3-dependent mitophagy in the neuroprotection of TAT-SPK2. Mitophagy was quantified by immunoblotting, immunofluorescence and electron microscopy. The interaction between TAT-SPK2 and Bcl-2, Bcl-2 and BNIP3 was detected by co-immunoprecipitation. In the tMCAO model, pre-treatment with TAT-SPK2 significantly reduced infarct volume, improved neurological function and decreased brain edema. Neuroprotection by TAT-SPK2 was still seen when the peptide was administered 3 h after reperfusion. TAT-SPK2 also significantly improved functional recovery and reduced long-term brain atrophy of the ischemic hemisphere 30 days after administration. Our studies further showed that TAT-SPK2 directly binds to Bcl-2 and disrupts Bcl-2/Beclin-1 or Bcl-2/BNIP3 complexes to induce mitophagy. These results suggest that TAT-SPK2 protects neurons against ischemia reperfusion injury by activating BNIP3-mediated mitophagy. Agents exploiting this molecular mechanism are potential candidates for the treatment of ischemic stroke.


Subject(s)
Gene Products, tat/pharmacology , Membrane Proteins/agonists , Mitochondrial Proteins/agonists , Mitophagy/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peptides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Reperfusion Injury/prevention & control , Animals , Autophagy , Beclin-1 , Brain Edema/prevention & control , Infarction, Middle Cerebral Artery/drug therapy , Ischemic Stroke/drug therapy , Male , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , RNA, Small Interfering/pharmacology
5.
Inflammation ; 41(4): 1498-1507, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29728804

ABSTRACT

Aseptic implant loosening is a devastating long-term complication of total joint arthroplasty. It is mainly initiated by the interaction of wear debris and macrophages. However, how does the chronic inflammation persist and how to stop it is poorly understood. Sphingosine kinases (SPHKs) are an essential feature of immunosuppressive M2 polarisation in macrophages and a promoter for chronic inflammation. In this study, RAW 264.7 macrophages were exposed to stimulation with titanium particles (0.1 mg/ml), and the subsequent expression of SPHKs and pro-inflammatory cytokines was evaluated. The effect of inhibitors of SPHKs (FTY720, PF543, and ABC294640) on titanium particle-challenged macrophages was analysed. As for results, the amount of sphingosine kinase (SPHK)-1 and SPHK-2 in RAW264.7 macrophages increased in the presence of titanium particles in a time-dependent manner. Two inhibitors of SPHKs (FTY720 and ABC294640) suppressed titanium particle-induced tumour necrosis factor (TNF)-α and interleukin (IL)-6 production in RAW264.7 macrophages. These findings suggest that persistent stimulation with titanium particles may lead to a consistent release of TNF-α and IL-6 via SPHK-2 activity, which may lead to aseptic implant loosening. Appropriate regulation of SPHK-2 may serve as a potential new strategy in the treatment of aseptic implant loosening.


Subject(s)
Inflammation/chemically induced , Interleukin-6/metabolism , Particulate Matter/adverse effects , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Titanium/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Mice , Prosthesis Failure/drug effects , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/drug effects
6.
Phytother Res ; 32(6): 1098-1107, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29468792

ABSTRACT

Alzheimer's disease (AD) is a very common neurodegenerative disorder in the elderly and brings considerable financial and social problems worldwide. In this study, polyprenols were firstly evaluated the effects on the cognitive deficits and neuropathology in APP/PS1 mice model of AD. At 3 months old, the APP/PS1 mice were divided into model group; polyprenols low, middle, and high dosage group; and positive drug group. Age-matched wild-type mice were chosen in control group. The administration by oral gavage lasted 6 months. Polyprenols treatment significantly improved cognitive impairment of double transgenic mice compared with vehicle control treatment in behavioral tests. In addition, immunohistochemistry and enzyme-linked immunosorbent assay showed that there were significantly reductions in neuritic plaques and the level of hyperphosphorylated tau in brain of polyprenols-treated mice. Furthermore, we found that polyprenols treatment reduced the apoptotic cells in brain sections of 9-month-old APP/PS1 mice. These results reveal that polyprenols exert neuroprotective effects in APP/PS1 mice and could represent an effective treatment for AD.


Subject(s)
Cognitive Dysfunction/drug therapy , Neuropathology/methods , Phosphotransferases (Alcohol Group Acceptor)/therapeutic use , Animals , Cognition Disorders/drug therapy , Cognitive Dysfunction/pathology , Disease Models, Animal , Humans , Male , Mice , Mice, Transgenic , Phosphotransferases (Alcohol Group Acceptor)/pharmacology
7.
J Nat Med ; 72(2): 390-398, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29260413

ABSTRACT

Renal ischemia reperfusion injury (RIRI) refers to the irreversible damage for renal function when blood perfusion is recovered after ischemia for an extended period, which is common in clinical surgeries and has been regarded as a major risk for acute renal failures (ARF) that is accompanied with unimaginably high morbidity and mortality. Hypoxia during ischemia followed by reoxygenation via reperfusion serves as a major event contributing to cell apoptosis, which has been widely accepted as the vital pathogenesis in RIRI. Preventing apoptosis in renal tubular epithelial cell has been considered as effective method for blocking RIRI. In this paper, we established a hypoxia/reoxygenation (H/R) injury model in human proximal tubular epithelial HK-2 cells. Here, we found increased SPHK1 levels in H/R injured HK-2 cells, which could be significantly down regulated after berberine treatment. Berberine has been reported to exert a protective effect on H/R-induced apoptosis of HK-2 cells. So, in our present study, we planned to investigate whether SPHK1 participated in the anti-apoptosis process of berberine in H/R injured HK-2 cells. Our study confirmed the protective effect of berberine against H/R-induced apoptosis in HK-2 cells through promoting cells viability, inhibiting cells apoptosis, and down-regulating p-P38, caspase-3, caspase-9 as well as SPHK1, while up regulating the ratio of Bcl-2/Bax. However, SPHK1 overexpression in HK-2 cells induced severe apoptosis, which can be significantly ameliorated with additional berberine treatment. We concluded that berberine could remarkably prevent H/R-induced apoptosis in HK-2 cells through down-regulating SPHK1 expression levels, and the mechanisms included the suppression of p38 MAPK activation and mitochondrial stress pathways.


Subject(s)
Berberine/therapeutic use , Caspase 3/metabolism , Caspase 9/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Animals , Apoptosis , Berberine/administration & dosage , Berberine/pharmacology , Cell Hypoxia , Cell Proliferation , Humans , Phosphotransferases (Alcohol Group Acceptor)/pharmacology
8.
Mol Oncol ; 11(5): 517-533, 2017 05.
Article in English | MEDLINE | ID: mdl-28258651

ABSTRACT

Strong experimental evidence in animal and cellular models supports a pivotal role of sphingosine kinase-1 (SK1) in oncogenesis. In many human cancers, SK1 levels are upregulated and these increases are linked to poor prognosis in patients. Here, by employing untargeted NMR-based metabolomic profiling combined with functional validations, we report the crucial role of SK1 in the metabolic shift known as the Warburg effect in A2780 ovarian cancer cells. Indeed, expression of SK1 induced a high glycolytic rate, characterized by increased levels of lactate along with increased expression of the proton/monocarboxylate symporter MCT1, and decreased oxidative metabolism, associated with the accumulation of intermediates of the tricarboxylic acid cycle and reduction in CO2 production. Additionally, SK1-expressing cells displayed a significant increase in glucose uptake paralleled by GLUT3 transporter upregulation. The role of SK1 is not limited to the induction of aerobic glycolysis, affecting metabolic pathways that appear to support the biosynthesis of macromolecules. These findings highlight the role of SK1 signaling axis in cancer metabolic reprogramming, pointing out innovative strategies for cancer therapies.


Subject(s)
Adenocarcinoma/metabolism , Glycolysis , Magnetic Resonance Imaging/methods , Metabolomics/methods , Ovarian Neoplasms/metabolism , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Adenocarcinoma/genetics , Animals , Carbon Dioxide/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Female , Glucose/metabolism , Glucose Transporter Type 3/metabolism , Glycolysis/drug effects , Humans , Lactic Acid/analysis , Lactic Acid/metabolism , Mitochondria/metabolism , Oncogene Proteins/metabolism , Ovarian Neoplasms/genetics , Oxidation-Reduction/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Prognosis , Tricarboxylic Acids/metabolism , Up-Regulation
9.
J Clin Pathol ; 69(9): 772-6, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26850632

ABSTRACT

AIMS: Human heart valves are prone to glycation, a fundamental process of ageing. The aim of this study was to establish the relationship between fructosamine formation and the mechanical properties of human aortic valves. METHODS: 67 patients (age: 76±8 years) diagnosed with an aortic valve stenosis, who underwent an aortic valve replacement were enrolled. Fructosamine and calcium concentrations in aortic valves were determined. Using a transthoracic Doppler echocardiography, aortic valve orifice area and transvalvular pressure gradients were measured. In a subgroup of 32 patients, the aortic valve orifice area was sufficient to carry out mechanical testing on a LFPlus Universal material tester. An in vitro removal of fructosamine of the valve was initiated using ATP-dependent fructosamine 3-kinase (FN3K). RESULTS: A significant correlation was found between the aortic valve fructosamine concentration and the calculated aortic valve orifice area: Y (aortic valve orifice area, mm(2))=1.050-0.228X (aortic valve fructosamine concentration, µmol/g valve) (r=-0.38). A significantly higher calcium concentration was measured in the aortic valves of diabetics in comparison with those of non-diabetics. A multiple regression analysis revealed that the presence of diabetes mellitus and aortic valve fructosamine concentration were the main predictors of the extensibility of the aortic valves. In the in vitro deglycation study, a significant lower aortic valve fructosamine concentration was detected after treatment with FN3K. This resulted in an increased flexibility of the aortic valves. CONCLUSIONS: Although no direct causativeness is proven with the presented results, which just show an association between fructosamine, the effect of FN3K and aortic valve stiffness, the present study points for the first time towards a possible additional role of the Amadori products in the biomechanical properties of ageing aortic valves.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Fructosamine/metabolism , Vascular Stiffness/physiology , Aged , Aged, 80 and over , Aortic Valve/diagnostic imaging , Aortic Valve/drug effects , Aortic Valve/pathology , Aortic Valve Stenosis/diagnostic imaging , Aortic Valve Stenosis/pathology , Calcium/metabolism , Echocardiography, Doppler , Female , Humans , Male , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Vascular Stiffness/drug effects
10.
Am J Pathol ; 180(4): 1740-50, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22322303

ABSTRACT

Leukocyte recruitment to sites of inflammation is critical for the development of acute allergic responses. Rapid P-selectin up-regulation by endothelial cells is a key promoter of leukocyte infiltration in response to mediators such as histamine. However, the mechanisms underpinning this process are still incompletely understood. We examined the role of the sphingosine kinase/sphingosine-1-phosphate (SK/S1P) pathway and showed that in human umbilical vein endothelial cells, histamine rapidly activates SK in an extracellular signal-regulated kinase (ERK) 1/2-dependent manner, concurrent with the induction of P-selectin expression. Histamine activated both SK-1 and SK-2 isoforms; inhibition of SK-1, but not SK-2, attenuated histamine-induced P-selectin up-regulation and neutrophil rolling in vitro. S1P receptor antagonists failed to prevent histamine-induced P-selectin expression, and exogenous S1P did not increase P-selectin expression, suggesting that S1P cell surface receptors are not involved in this process. Finally, the role of both SK-1 and SK-2 in histamine-induced leukocyte rolling in vivo was assessed using pharmacological and genetic methods. Consistent with the in vitro findings, mice pretreated with either sphingosine kinase inhibitor or fingolimod (FTY720) significantly attenuated histamine-induced leukocyte rolling in the cremaster muscle. Similarly, Sphk1(-/-) but not Sphk2(-/-) mice exhibited reduced histamine-induced leukocyte rolling. These findings demonstrate a key role for SK-1 in histamine-induced rapid P-selectin up-regulation and associated leukocyte rolling, and suggest that endothelial SK-1 is an important contributor to allergic inflammation.


Subject(s)
Histamine/pharmacology , Neutrophil Infiltration/drug effects , Phosphotransferases (Alcohol Group Acceptor)/physiology , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fingolimod Hydrochloride , Hemodynamics/physiology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunosuppressive Agents/pharmacology , Leukocyte Count , Leukocyte Rolling/drug effects , Leukocyte Rolling/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Neutrophil Infiltration/physiology , P-Selectin/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/physiology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Up-Regulation/drug effects
11.
Plant J ; 65(3): 453-68, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21265898

ABSTRACT

The regulation of pollen tube growth by the phospholipid phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P(2) ) is not well understood. The Arabidopsis genome encodes two type A phosphatidylinositol-4-phosphate (PI4P) 5-kinases, PIP5K10 and PIP5K11, which are exclusively expressed in pollen and produce PtdIns(4,5)P(2) in vitro. Fluorescence-tagged PIP5K10 and PIP5K11 localized to lateral subapical plasma membrane microdomains in tobacco pollen tubes in a pattern closely resembling the distribution of PtdIns(4,5)P(2,) with the exception of notably weaker association at the extreme apex. Overexpression of PIP5K10 or PIP5K11 in tobacco pollen tubes resulted in severe tip swelling and altered actin fine structure similar to that reported for overexpression of tobacco Nt-Rac5, a monomeric GTPase known to regulate the actin cytoskeleton. Increased sensitivity of Arabidopsis pip5k10 pip5k11 double mutant pollen tubes to Latrunculin B (LatB) further supports a role for type A PI4P 5-kinases in controlling the actin cytoskeleton. Despite the disruption of both its type A PI4P 5-kinases, the pip5k10 pip5k11 double mutant was fertile, indicating that one of the remaining type B PI4P 5-kinase isoforms might be functionally redundant with PIP5K10 and PIP5K11. Antagonistic effects of PIP5K11 and the Nt-Rac5-specific guanine nucleotide dissociation inhibitor, Nt-RhoGDI2, on tip swelling observed in coexpression-titration experiments indicate a link between PtdIns(4,5)P(2) and Rac-signaling in pollen tubes. The data suggest that type A PI4P 5-kinases influence the actin cytoskeleton in pollen tubes in part by counteracting Nt-RhoGDI2, possibly contributing to the control of the pool of plasma membrane-associated Nt-Rac5.


Subject(s)
Arabidopsis/growth & development , Nicotiana/growth & development , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pollen Tube/growth & development , rac GTP-Binding Proteins/metabolism , Actins/chemistry , Arabidopsis/drug effects , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Membrane/metabolism , DNA, Bacterial/genetics , Guanine Nucleotide Dissociation Inhibitors/genetics , Guanine Nucleotide Dissociation Inhibitors/metabolism , Mutagenesis, Insertional , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Plant Proteins/genetics , Plant Proteins/physiology , Plants, Genetically Modified/genetics , Plants, Genetically Modified/growth & development , Plants, Genetically Modified/metabolism , Pollen Tube/genetics , Pollen Tube/metabolism , Thiazolidines/pharmacology , Nicotiana/genetics , Nicotiana/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/genetics
12.
ACS Chem Biol ; 5(2): 223-32, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20025244

ABSTRACT

Bacterial quorum sensing (QS) is a cell-cell communication process, mediated by signaling molecules, that alters various phenotypes including pathogenicity. Methods to interrupt these communication networks are being pursued as next generation antimicrobials. We present a technique for interrupting communication among bacteria that exploits their native and highly specific machinery for processing the signaling molecules themselves. Specifically, our approach is to bring native intracellular signal processing mechanisms to the extracellular surroundings and "quench" crosstalk among a variety of strains. In this study, the QS system based on the interspecies signaling molecule autoinducer-2 (AI-2) is targeted because of its prevalence among prokaryotes (it functions in over 80 bacterial species). We demonstrate that the Escherichia coli AI-2 kinase, LsrK, can phosphorylate AI-2 in vitro, and when LsrK-treated AI-2 is added ex vivo to E. coli populations, the native QS response is significantly reduced. Further, LsrK-mediated degradation of AI-2 attenuates the QS response among Salmonella typhimurium and Vibrio harveyi even though the AI-2 signal transduction mechanisms and the phenotypic responses are species-specific. Analogous results are obtained from a synthetic ecosystem where three species of bacteria (enteric and marine) are co-cultured. Finally, the addition of LsrK and ATP to growing co-cultures of E. coli and S. typhimurium exhibits significantly reduced native "cross-talk" that ordinarily exists among and between species in an ecosystem. We believe this nature-inspired enzymatic approach for quenching QS systems will spawn new methods for controlling cell phenotype and potentially open new avenues for controlling bacterial pathogenicity.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Homoserine/analogs & derivatives , Lactones/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Quorum Sensing , Coculture Techniques , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli Proteins/pharmacology , Homoserine/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Salmonella typhimurium/drug effects , Salmonella typhimurium/enzymology , Salmonella typhimurium/growth & development , Vibrio/drug effects , Vibrio/enzymology , Vibrio/growth & development
13.
Biochemistry ; 45(9): 2968-77, 2006 Mar 07.
Article in English | MEDLINE | ID: mdl-16503651

ABSTRACT

Hereditary inclusion body myopathy (HIBM), a neuromuscular disorder, is caused by mutations in UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE), the key enzyme of sialic acid biosynthesis. To date, more than 40 different mutations in the GNE gene have been reported to cause the disease. Ten of them, representing mutations in both functional domains of GNE, were recombinantly expressed in insect cells (Sf9). Each of the mutants that was analyzed displayed a reduction in the two known GNE activities, thus revealing that mutations may also influence the function of the domain not harboring them. The extent of reduction strongly differs among the point mutants, ranging from only 20% reduction found for A631T and A631V to almost 80% reduction of at least one activity in D378Y and N519S mutants and more than 80% reduction of both activities of G576E, underlined by structural changes of N519S and G576E, as observed in CD spectroscopy and gel filtration analysis, respectively. We therefore generated models of the three-dimensional structures of the epimerase and the kinase domains of GNE, based on Escherichia coli UDP-N-acetylglucosamine 2-epimerase and glucokinase, respectively, and determined the localization of the HIBM mutations within these proteins. Whereas in the kinase domain most of the mutations are localized inside the enzyme, mutations in the epimerase domain are mostly located at the protein surface. Otherwise, the different mutations result in different enzymatic activities but not in different disease phenotypes and, therefore, do not suggest a direct role of the enzymatic function of GNE in the disease mechanism.


Subject(s)
Myositis, Inclusion Body/metabolism , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Baculoviridae/metabolism , Carbohydrate Epimerases/genetics , Carbohydrate Epimerases/metabolism , Carbohydrate Epimerases/pharmacology , Humans , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Structure, Tertiary/genetics , Protein Transport , Structure-Activity Relationship
14.
Cancer Chemother Pharmacol ; 58(4): 547-54, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16463058

ABSTRACT

PURPOSE: Cytotoxic nucleoside analogues are widely used in the treatment of cancers. Resistance to these compounds is frequent and often multifactorial. Deficiency in deoxycytidine kinase (dCK), the rate-limiting activating enzyme, has been reported in a number of in vitro models as well as in various clinical situations. Some strategies to overcome this mechanism of resistance have been proposed there by gene transfer based therapy. METHODS: We have developed and characterized a gemcitabine-resistant cell line (Messa 10 K) from the human uterine sarcoma Messa strain, and transfected this cell line with the multisubstrate deoxynucleoside kinase from Drosophila melanogaster (DmdNK) in order to revert the resistance in Messa 10 K cells which was due to dCK-deficiency. RESULTS: Messa 10 K is highly resistant to gemcitabine (122-fold), troxacitabine (>15-fold) and araC (13,556-fold). Quantitative real-time PCR and western blot analysis showed that dCK was not detectable in Messa 10 K cells, presumably because of a genetic modification. The transfection of Messa 10 K cells with DmdNK significantly increased the sensitivity to gemcitabine. CONCLUSIONS: These results show that genetic modifications in non-hematological malignant cells may be associated with resistance to gemcitabine, and that the gene transfer of non-human genes can be used for the reversion of nucleoside analogue resistance due to dCK deficiency.


Subject(s)
Antineoplastic Agents/pharmacology , DNA, Neoplasm/genetics , Deoxycytidine Kinase/deficiency , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/metabolism , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Sarcoma/enzymology , Animals , Cell Line, Tumor , Deoxycytidine/metabolism , Drosophila melanogaster , Humans , Sarcoma/genetics , Transfection/methods , Gemcitabine
15.
J Biol Chem ; 281(13): 8518-27, 2006 Mar 31.
Article in English | MEDLINE | ID: mdl-16415355

ABSTRACT

The sphingolipid ceramide induces macroautophagy (here called autophagy) and cell death with autophagic features in cancer cells. Here we show that overexpression of sphingosine kinase 1 (SK1), an enzyme responsible for the production of sphingosine 1-phosphate (S1P), in MCF-7 cells stimulates autophagy by increasing the formation of LC3-positive autophagosomes and the rate of proteolysis sensitive to the autophagy inhibitor 3-methyladenine. Autophagy was blocked in the presence of dimethylsphingosine, an inhibitor of SK activity, and in cells expressing a catalytically inactive form of SK1. In SK1(wt)-overexpressing cells, however, autophagy was not sensitive to fumonisin B1, an inhibitor of ceramide synthase. In contrast to ceramide-induced autophagy, SK1(S1P)-induced autophagy is characterized by (i) the inhibition of mammalian target of rapamycin signaling independently of the Akt/protein kinase B signaling arm and (ii) the lack of robust accumulation of the autophagy protein Beclin 1. In addition, nutrient starvation induced both the stimulation of autophagy and SK activity. Knocking down the expression of the autophagy protein Atg7 or that of SK1 by siRNA abolished starvation-induced autophagy and increased cell death with apoptotic hallmarks. In conclusion, these results show that SK1(S1P)-induced autophagy protects cells from death with apoptotic features during nutrient starvation.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cell Survival/drug effects , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Starvation , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 7 , Beclin-1 , Blotting, Western , Breast Neoplasms/pathology , Cell Line, Tumor , Ceramides/analysis , Enzyme Inhibitors/pharmacology , Female , Green Fluorescent Proteins/metabolism , Humans , Hydrolysis , Lactosylceramides/metabolism , Membrane Proteins/metabolism , Phospholipase D/analysis , Phosphotransferases (Alcohol Group Acceptor)/analysis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , TOR Serine-Threonine Kinases , Ubiquitin-Activating Enzymes/metabolism
16.
Leukemia ; 20(1): 95-102, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16281067

ABSTRACT

We examined the involvement of sphingosine kinase-1, a critical regulator of the sphingolipid balance, in susceptibility to antineoplastic agents of either sensitive or multidrug-resistant acute myeloid leukemia cells. Contrary to parental HL-60 cells, doxorubicin and etoposide failed to trigger apoptosis in chemoresistant HL-60/Doxo and HL-60NP16 cells overexpressing MRP1 and MDR1, respectively. Chemosensitive HL-60 cells displayed sphingosine kinase-1 inhibition coupled with ceramide generation. In contrast, chemoresistant HL-60/ Doxo and HL-60/VP16 had sustained sphingosine kinase-1 activity and did not produce ceramide during treatment. Enforced expression of sphingosine kinase-1 in chemosensitive HL-60 cells resulted in marked inhibition of apoptosis that was mediated by blockade of mitochondrial cytochrome c efflux hence suggesting a control of apoptosis at the pre-mitochondrial level. Incubation with cell-permeable ceramide of chemoresistant cells led to a sphingosine kinase-1 inhibition and apoptosis both prevented by sphingosine kinase-1 over-expression. Furthermore, F-12509a, a new sphingosine kinase inhibitor, led to ceramide accumulation, decrease in sphingosine 1-phosphate content and caused apoptosis equally in chemosensitive and chemoresistant cell lines that is inhibited by adding sphingosine 1-phosphate or overexpressing sphingosine kinase-1. F-12509a induced classical apoptosis hallmarks namely nuclear fragmentation, caspase-3 cleavage as well as downregulation of antiapoptotic XIAP, and release of cytochrome c and SMAC/Diablo.


Subject(s)
Drug Resistance, Multiple , Leukemia, Myeloid/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Acute Disease , Apoptosis/drug effects , Benzoquinones/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Ceramides/biosynthesis , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Etoposide/pharmacology , HL-60 Cells , Humans , Leukemia, Myeloid/drug therapy , Mitochondria/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , RNA Interference/physiology , Receptors, Lysosphingolipid/metabolism
17.
J Leukoc Biol ; 77(2): 181-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15522918

ABSTRACT

Lysophosphatidic acid (LPA) is a bioactive lipid, which is structurally similar to sphingosine 1-phosphate (S1P) and which can mobilize Ca2+ in multiple cell types. We recently showed that S1P induces Ca2+ entry directly through store-operated Ca2+ entry (SOCE) channels in human polymorphonuclear neutrophils (PMN). We therefore examined the mechanisms by which LPA induces intracellular Ca2+ mobilization in PMN. External application of low micromolar LPA caused dose-dependent Ca2+ influx without releasing Ca2+ stores, whereas G-protein-coupled (GPC) LPA receptors respond to nanomolar LPA. Additive Ca2+ influx by LPA compared with 100 nM ionomycin-induced Ca2+ influx suggests that LPA-induced Ca2+ influx does not pass through SOCE channels. Ca2+ influx was resistant to inhibition of Gi/o by pertussis toxin, of phospholipase C by U73122, and of G12/13/Rho by Y27632, all demonstrating GPC receptor independence. This Ca2+ influx was inhibited by Gd3+, La3+, Zn2+, or MRS1845 but not by Ni2+ or the sphingosine kinase inhibitor dimethylsphingosine. In addition, we found that LPA has no effect on neutrophil chemotaxis; however, it has stimulatory effects on neutrophil respiratory burst in a dose-response manner. These findings suggest that LPA-induced Ca2+ influx in PMN occurs through a mechanism other than SOCE channels, independent of Ca2+ store-depletion and S1P synthesis, and that the characteristics of LPA-induced Ca2+ influx are similar to those of S1P-induced influx in terms of sensitivity to inorganic inhibitors. Unlike S1P, LPA has stimulatory effects on neutrophil respiratory burst.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Lysophospholipids/metabolism , Neutrophils/metabolism , Sphingosine/analogs & derivatives , Calcium/antagonists & inhibitors , Calcium Signaling/drug effects , Chemotaxis/physiology , Estrenes/pharmacology , HL-60 Cells , Humans , Ionomycin/pharmacology , Lysophospholipids/pharmacology , Models, Biological , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Pyrrolidinones/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Respiratory Burst/drug effects , Respiratory Burst/physiology , Signal Transduction/physiology , Sphingosine/metabolism
18.
Skin Pharmacol Physiol ; 17(2): 77-83, 2004.
Article in English | MEDLINE | ID: mdl-14976384

ABSTRACT

Hyaluronan (HA) is well known to reside in the extracellular matrix as a water-sorbed macromolecule. The aims of this study were twofold: to investigate the regulation of HA synthesis in keratinocytes, and to develop a method to modulate this regulatory process. We found that N-acetylglucosamine (NAG) increased the production of HA by cultured keratinocytes dose dependently, but had no effect on the production by skin fibroblasts. The effect of NAG in keratinocytes was found to be specific for HA production, as there was no change in sulfated glycosaminoglycan formation. The copresence of NAG with either of two retinoids, retinoic acid (RA) or retinol, exerted a synergistic effect on HA production. To investigate whether human HA synthase (HAS) genes were regulated by NAG or retinoids, total RNA extracted from cells treated with these agents was subjected to Northern blot analysis. We observed that RA and retinol markedly induced the expression of HA synthase-3 (HAS3) mRNA. Moreover, beta-carotene, a provitamin A, influenced HA production and HAS3 gene expression in a manner similar to the retinoids. Conversely, NAG had no effect on the expression of HAS3 transcripts. Pretreatment of cells with RA stimulated the activity of membrane-associated HAS, whereas pretreatment with NAG did not. These results suggest that HA production is regulated by at least two pathways: one involving the regulation of HAS gene expression, and the other independent of such a regulatory effect. Taken together, our findings suggest that NAG is a new modulator of HA synthesis.


Subject(s)
Drug Synergism , Hyaluronic Acid/biosynthesis , Keratinocytes/metabolism , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Retinoids/pharmacology , Animals , Blotting, Northern/methods , Cattle , Cell Culture Techniques , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Gene Expression/drug effects , Glucuronosyltransferase/drug effects , Glucuronosyltransferase/genetics , Humans , Hyaluronan Synthases , Keratinocytes/drug effects , Male , Phosphotransferases (Alcohol Group Acceptor)/physiology , RNA, Messenger , Retinoids/chemistry , Transferases/metabolism , beta Carotene/pharmacology
19.
J Biol Chem ; 278(47): 46452-60, 2003 Nov 21.
Article in English | MEDLINE | ID: mdl-12963721

ABSTRACT

Sphingosine 1-phosphate (S1P) is the ligand for a family of specific G protein-coupled receptors (GPCRs) that regulate a wide variety of important cellular functions, including growth, survival, cytoskeletal rearrangements, and cell motility. However, whether it also has an intracellular function is still a matter of great debate. Overexpression of sphingosine kinase type 1, which generated S1P, induced extensive stress fibers and impaired formation of the Src-focal adhesion kinase signaling complex, with consequent aberrant focal adhesion turnover, leading to inhibition of cell locomotion. We have dissected biological responses dependent on intracellular S1P from those that are receptor-mediated by specifically blocking signaling of Galphaq, Galphai, Galpha12/13, and Gbetagamma subunits, the G proteins that S1P receptors (S1PRs) couple to and signal through. We found that intracellular S1P signaled "inside out" through its cell-surface receptors linked to G12/13-mediated stress fiber formation, important for cell motility. Remarkably, cell growth stimulation and suppression of apoptosis by endogenous S1P were independent of GPCRs and inside-out signaling. Using fibroblasts from embryonic mice devoid of functional S1PRs, we also demonstrated that, in contrast to exogenous S1P, intracellular S1P formed by overexpression of sphingosine kinase type 1 promoted growth and survival independent of its GPCRs. Hence, exogenous and intracellularly generated S1Ps affect cell growth and survival by divergent pathways. Our results demonstrate a receptor-independent intracellular function of S1P, reminiscent of its action in yeast cells that lack S1PRs.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/physiology , Lysophospholipids , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Sphingosine/analogs & derivatives , Stress Fibers/metabolism , Animals , Cell Division , Cell Movement , Cells, Cultured , Embryo, Mammalian , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Focal Adhesions/metabolism , Mice , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein-Tyrosine Kinases , Receptors, G-Protein-Coupled , Signal Transduction , Sphingosine/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...