Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 22(10): 1676-86, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25744023

ABSTRACT

Neuronal active Caspase-6 (Casp6) is associated with Alzheimer disease (AD), cognitive impairment, and axonal degeneration. Caspase-1 (Casp1) can activate Casp6 but the expression and functionality of Casp1-activating inflammasomes has not been well-defined in human neurons. Here, we show that primary cultures of human CNS neurons expressed functional Nod-like receptor protein 1 (NLRP1), absent in melanoma 2, and ICE protease activating factor, but not the NLRP3, inflammasome receptor components. NLRP1 neutralizing antibodies in a cell-free system, and NLRP1 siRNAs in neurons hampered stress-induced Casp1 activation. NLRP1 and Casp1 siRNAs also abolished stress-induced Casp6 activation in neurons. The functionality of the NLRP1 inflammasome in serum-deprived neurons was also demonstrated by NLRP1 siRNA-mediated inhibition of speck formation of the apoptosis-associated speck-like protein containing a caspase recruitment domain conjugated to green fluorescent protein. These results indicated a novel stress-induced intraneuronal NLRP1/Casp1/Casp6 pathway. Lipopolysaccharide induced Casp1 and Casp6 activation in wild-type mice brain cortex, but not in that of Nlrp1(-/-) and Casp1(-/-) mice. NLRP1 immunopositive neurons were increased 25- to 30-fold in AD brains compared with non-AD brains. NLRP1 immunoreactivity in these neurons co-localized with Casp6 activity. Furthermore, the NLRP1/Casp1/Casp6 pathway increased amyloid beta peptide 42 ratio in serum-deprived neurons. Therefore, CNS human neurons express functional NLRP1 inflammasomes, which activate Casp1 and subsequently Casp6, thus revealing a fundamental mechanism linking intraneuronal inflammasome activation to Casp1-generated interleukin-1-ß-mediated neuroinflammation and Casp6-mediated axonal degeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Caspase 6/metabolism , Inflammasomes/metabolism , Interleukin-1beta/biosynthesis , Neurons/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Axons/metabolism , Cells, Cultured , Humans , Inflammation , Male , Mice , NLR Proteins , Neurons/physiology , Signal Transduction
2.
Clin Exp Allergy ; 41(2): 260-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20718780

ABSTRACT

BACKGROUND: Studies in both human and mouse indicate that mediators released by mast cells can lead to bronchoconstriction, and thus these are important effector cells in lifethreatening anaphylaxis. Much of our understanding of the various functions of mast cells emanates from the study of mice lacking these cells, particularly mice carrying mutations in the tyrosine kinase gene Kit. Definitive evidence for the role of mast cells in the altered immune response requires the demonstration that this response can be normalized by reconstitution of the mice with cultured bone marrow-derived mast cells (BMMCs). While many mast cell niches can be restored with BMMCs, this has not been demonstrated for mast cells present in the airways of the lung, cells poised to mediate bronchoconstriction during allergic responses. OBJECTIVE: To determine if mast cell-deficient Kit(Wsh/Wsh) reconstituted lines are an appropriate model for the study of the role of these cells in bronchoconstriction associated with allergic responses. METHODS: Kit(Wsh/Wsh) mice were reconstituted with either whole bone marrow (WBM) or BMMCs and responses to IgE-mediated mast cell activation were determined; including systemic hypothermia, mediator release, and bronchoconstriction in anaesthetized, mechanically ventilated animals. RESULTS: Engraftment of Kit(Wsh/Wsh) mice with WBM and BMMCs results in reconstitution of the central airways with mast cells. While the treatment of the two groups of animals resulted in systemic changes when challenged with IgE/Ag in a model of passive anaphylaxis, bronchoconstriction was observed only in kit(Wsh/Wsh) animals, which had received a bone marrow transplant. CONCLUSIONS: While BMMCs can populate the lung, they cannot restore IgE/Ag-mediated bronchoconstriction to mast cell-deficient animals. This suggests that the mast cell population, which mediates this function, may be unique, and to fill this niche in the lung cells must undergo a specific developmental programme, one that is no longer available to cultured mast cells.


Subject(s)
Antigens/immunology , Bronchoconstriction/immunology , Lung/cytology , Lung/immunology , Mast Cells/immunology , Animals , Inflammation , Mice , Mice, Inbred C57BL , Neurons/immunology
3.
J Neural Transm (Vienna) ; 114(7): 899-908, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17318306

ABSTRACT

Induction of Fos protein by the potent and direct NMDA agonist (tetrazol-5-yl)glycine (TZG) was examined in mice. Effects of antipsychotic drugs were assessed on this in vivo index of NMDA receptor activation. TZG induced the expression of Fos in a neuroanatomically selective manner, with the hippocampal formation showing the most robust response. In mice genetically altered to express low levels of the NR1 subunit of the NMDA receptor, TZG-induced Fos was reduced markedly in comparison to the wild type controls. TZG-induced Fos was also blocked by the selective NMDA antagonist MK-801. Pretreatment of mice with clozapine (3 and 10 mg/kg) reduced TZG-induced Fos in the hippocampal formation but not in other brain regions. Haloperidol at a dose of 0.5 mg/kg did not antagonize TZG induced Fos in any region. Haloperidol at a dose of 1.0 mg/kg did attenuate the induction of Fos by TZG in the hippocampus but not in other brain regions. The relatively high dose (1 mg/kg) of haloperidol required to block effects of TZG suggests that this action may not be related to the D(2) dopamine receptor-blocking properties, since maximal D(2) receptor blockade was probably achieved by the 0.5 mg/kg dose of haloperidol. The antidepressant drug imipramine (10 or 20 mg/kg) did not antagonize TZG induced Fos in any brain region. The data suggest that clozapine can reduce excessive activation of NMDA receptors by TZG administration in vivo at doses relevant to the drugs' actions in rodent models of antipsychotic activity. Whether or not this action of clozapine contributes to its therapeutic properties will require further study.


Subject(s)
Clozapine/pharmacology , Glycine/analogs & derivatives , Haloperidol/pharmacology , N-Methylaspartate/agonists , Proto-Oncogene Proteins c-fos/biosynthesis , Receptors, N-Methyl-D-Aspartate/metabolism , Tetrazoles/pharmacology , Animals , Glycine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-fos/genetics , Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/genetics
4.
Ocul Immunol Inflamm ; 14(3): 157-63, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16766399

ABSTRACT

PURPOSE: To examine the role of EP2 and EP4 receptors in murine ocular inflammation. METHODS: Prostaglandin EP2 and EP4 receptor knockout and wild-type mice were treated topically with prostaglandin E2, SDF-1, and RANTES and lipopolysaccharide by intravitreal injection. Paracentesis was performed by puncturing the cornea. The increase in the level of aqueous humor protein and the number of leukocytes were measured and the vascular leakage of protein was visualized using fluorescein angiography. RESULTS: In the EP2 receptor knockout mice, there was significant inhibition of the disruption of the blood-aqueous barrier caused by lipopolysaccharides, paracentesis, prostaglandin E2, SDF-1, and RANTES. Reductions in the disruption in the blood-aqueous barrier and leukocyte infiltration after lipopolysaccharide injection and paracentesis were significant, but there was no increase in the aqueous humor protein level after prostaglandin E2 treatment in EP4 receptor knockout mice. CONCLUSIONS: The results of the present experiments suggest that EP2 and EP4 receptors partly mediate the disruption of the blood-aqueous barrier and leukocyte infiltration induced by prostaglandin E2, SDF-1, RANTES, and lipopolysaccharides.


Subject(s)
Aqueous Humor/metabolism , Receptors, Prostaglandin E/physiology , Uveitis, Anterior/immunology , Animals , Anterior Chamber , Biomarkers/metabolism , Blood-Aqueous Barrier , Disease Models, Animal , Fluorescein Angiography , Fundus Oculi , Injections , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Uveitis, Anterior/chemically induced , Uveitis, Anterior/pathology
5.
J Immunol ; 167(11): 6631-6, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11714834

ABSTRACT

Acute renal allograft rejection is associated with alterations in renal arachidonic acid metabolism, including enhanced synthesis of leukotrienes (LTs). LTs, the products of the 5-lipoxygenase (5-LO) pathway, are potent lipid mediators with a broad range of biologic activities. Previous studies, using pharmacological agents to inhibit LT synthesis or activity, have implicated these eicosanoids in transplant rejection. To further investigate the role of LTs in acute graft rejection, we transplanted kidneys from CByD2F1 mice into fully allogeneic 129 mice that carry a targeted mutation in the 5lo gene. Unexpectedly, allograft rejection was significantly accelerated in 5-LO-deficient mice compared with wild-type animals. Despite the marked reduction in graft survival, the 5lo mutation had no effect on the hemodynamics or morphology of the allografts. Although LTB4 levels were reduced, renal thromboxane B2 production and cytokine expression were not altered in 5-LO-deficient allograft recipients. These findings suggest that, along with their proinflammatory actions, metabolites of 5-LO can act to enhance allograft survival.


Subject(s)
Arachidonate 5-Lipoxygenase/deficiency , Arachidonate 5-Lipoxygenase/genetics , Graft Rejection/enzymology , Graft Rejection/genetics , Kidney Transplantation/immunology , Lipoxins , Animals , Arachidonate 5-Lipoxygenase/metabolism , Crosses, Genetic , Cytokines/biosynthesis , Cytokines/genetics , Graft Rejection/physiopathology , Hydroxyeicosatetraenoic Acids/biosynthesis , Kidney Function Tests , Kidney Transplantation/pathology , Leukotriene B4/biosynthesis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Knockout , RNA, Messenger/biosynthesis , Thromboxane A2/biosynthesis , Thromboxane B2/biosynthesis
6.
J Clin Invest ; 108(8): 1229-35, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11602631

ABSTRACT

Production of prostaglandin E(2) (PGE(2)) is enhanced during inflammation, and this lipid mediator can dramatically modulate immune responses. There are four receptors for PGE(2) (EP1-EP4) with unique patterns of expression and different coupling to intracellular signaling pathways. To identify the EP receptors that regulate cellular immune responses, we used mouse lines in which the genes encoding each of the four EP receptors were disrupted by gene targeting. Using the mixed lymphocyte response (MLR) as a model cellular immune response, we confirmed that PGE(2) has potent antiproliferative effects on wild-type responder cells. The absence of either the EP1 or EP3 receptors did not alter the inhibitory response to PGE(2) in the MLR. In contrast, when responder cells lacked the EP2 receptor, PGE(2) had little effect on proliferation. Modest resistance to PGE(2) was also observed in EP4-/- responder cells. Reconstitution experiments suggest that EP2 receptors primarily inhibit the MLR through direct actions on T cells. Furthermore, PGE(2) modulates macrophage function by activating the EP4 receptor and thereby inhibiting cytokine release. Thus, PGE(2) regulates cellular immune responses through distinct EP receptors on different immune cell populations: EP2 receptors directly inhibit T cell proliferation while EP2 and EP4 receptors regulate antigen presenting cells functions.


Subject(s)
Immunity, Cellular , Receptors, Prostaglandin E/immunology , Animals , Antigen-Presenting Cells/immunology , Base Sequence , DNA Primers/genetics , Dinoprostone/pharmacology , Gene Expression , Interleukin-12/biosynthesis , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred DBA , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prostaglandin E/classification , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP3 Subtype , Receptors, Prostaglandin E, EP4 Subtype , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/biosynthesis
7.
Cancer Res ; 61(17): 6577-82, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11522657

ABSTRACT

Loss of p53 function is known to compromise cell cycle regulation, inductionof apoptosis, and DNA damage repair and can facilitate neoplastic transformation of cells. Mutations in the p53 gene are identified frequently in breast carcinomas. Li-Fraumeni patients inheriting a mutant p53 allele have an increased risk for developing tumors including breast cancer. Although mouse lines carrying mutations in the p53 gene have been generated, they die primarily of lymphoma and thus to date provide a limited model for the study of this disease and the role of p53 in nonfamilial breast cancer. An increasing body of literature suggests that the incidence of various tumors is determined largely by the genetic background on which mutations are studied. In addition, population studies and studies in animals suggest that environmental factors, together with genetic factors, determine overall risk for development of specific types of tumors. We therefore examined the impact of genetic background together with exposure to ionizing radiation on the development of tumors, particularly mammary tumors, in p53-deficient animals. We report here that modifier alleles present in the BALB/c strain increase the incidence of hemangiosarcomas [15 of 53 (28.3%); P = 0.0007] in p53(-/-) mice above rates reported previously in p53(-/-) mice on a mixed background as compared to the incidence observed in DBA/p53(-/-) mice. However, no increase in the frequency of mammary tumors is seen in these mice or in p53(-/-) DBA/2 animals, nor was an increase in mammary tumors observed in the DBA/2 p53(+/-) mice, even after exposure to 5 Gy of whole-body ionizing radiation. In contrast, a significant increase in the incidence of mammary tumors was observed in similarly treated BALB/c p53(+/-) mice (37.3% versus 6.8%; P = 0.0007). This was accompanied by a comparable decrease in the incidence of lymphomas. These results show that environmental agents together with genetic factors can increase the frequency and decrease the latency of mammary tumors, leading to an incidence similar to that observed in Li-Fraumeni syndrome. Furthermore, it suggests that the risk of development of a particular type of tumor by individuals deficient in p53 after exposure to damaging agents can be influenced by modifier alleles.


Subject(s)
Cocarcinogenesis , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/genetics , Neoplasms, Radiation-Induced/genetics , Tumor Suppressor Protein p53/deficiency , Alleles , Animals , Crosses, Genetic , DNA/radiation effects , DNA Damage , Female , Genes, p53/genetics , Genetic Predisposition to Disease , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Neoplasms, Radiation-Induced/etiology , Tumor Suppressor Protein p53/genetics , Whole-Body Irradiation/adverse effects
8.
Am J Respir Cell Mol Biol ; 25(1): 14-20, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11472970

ABSTRACT

Chloride (Cl(-)) movement across fetal lung epithelia is thought to be mediated by the sodium-potassium-2-Cl(-) cotransporter NKCC1. We studied the role of NKCC1 in Cl(-) and liquid secretion in late-gestation NKCC-null (-/-) and littermate control fetal mouse lung. NKCC -/- mice had decreased lung water compared with littermate controls (wet/dry: control, 8.01 +/- 0.09; NKCC -/-, 7.06 +/- 0.14). Liquid secretion by 17-d NKCC -/- distal lung explants was similar to control explants. Bumetanide inhibited basal liquid secretion in control but not NKCC -/- explants (expansion over 48 h: control, 35 +/- 4%; NKCC -/- 46 +/- 7%). Treatment with 4,4'-diisothiocyanto-stilbene-2,2'-disulfonic acid (DIDS) decreased liquid secretion in both control and NKCC -/- explants. Basal transepithelial potential difference (PD) of control tracheal explants was higher than that of NKCC -/- (control, -13.7 +/- 0.5 mV; NKCC -/-, -11.6 +/- 0.6 mV). Amiloride (10(-)(4) M) inhibited basal PD to the same extent in control and NKCC -/- mice. Terbutaline-stimulated hyperpolarization was less in NKCC -/- than in control tracheas (DeltaPD: control, -10.8 +/- 1.33 mV; NKCC -/-, -6.1 +/- 0.7 mV) and was inhibited by DIDS and acetazolamide in NKCC -/- but not wild-type explants. We conclude that NKCC is rate-limiting for transcellular Cl(-) transport, and that alternative anion transport mechanisms can sustain liquid production at near-normal levels in the fetal NKCC -/- mouse lung.


Subject(s)
Carrier Proteins/physiology , Chlorides/metabolism , Lipid Metabolism , Lung/metabolism , Trachea/metabolism , Animals , Carbonates/metabolism , Carrier Proteins/genetics , Embryonic and Fetal Development , Epithelial Cells/metabolism , Genotype , Hydrogen-Ion Concentration , In Situ Hybridization , In Vitro Techniques , Ion Transport , Lung/cytology , Lung/embryology , Membrane Potentials , Mice , Sodium-Potassium-Chloride Symporters , Trachea/cytology , Trachea/embryology
9.
Am J Physiol Cell Physiol ; 281(2): C615-23, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11443061

ABSTRACT

Airways of Na(+)-K(+)-2Cl(-) (NKCC1)-deficient mice (-/-) were studied in Ussing chambers to determine the role of the basolateral NKCC1 in transepithelial anion secretion. The basal short-circuit current (I(sc)) of tracheae and bronchi from adult mice did not differ between NKCC1-/- and normal mice, whereas NKCC1-/- tracheae from neonatal mice exhibited a significantly reduced basal I(sc). In normal mouse tracheae, sensitivity to the NKCC1 inhibitor bumetanide correlated inversely with the age of the mouse. In contrast, tracheae from NKCC1-/- mice at all ages were insensitive to bumetanide. The anion secretory response to forskolin did not differ between normal and NKCC1-/- tissues. However, when larger anion secretory responses were induced with UTP, airways from the NKCC1-/- mice exhibited an attenuated response. Ion substitution and drug treatment protocols suggested that HCO secretion compensated for reduced Cl(-) secretion in NKCC1-/- airway epithelia. The absence of spontaneous airway disease or pathology in airways from the NKCC1-/- mice suggests that the NKCC1 mutant mice are able to compensate adequately for absence of the NKCC1 protein.


Subject(s)
Carrier Proteins/physiology , Trachea/metabolism , Aging/physiology , Animals , Animals, Newborn/growth & development , Animals, Newborn/physiology , Biological Transport/physiology , Bronchi/metabolism , Bumetanide/pharmacology , Carrier Proteins/genetics , Electric Conductivity , In Vitro Techniques , Ions , Mice , Mice, Knockout/genetics , Reference Values , Sodium-Potassium-Chloride Symporters , Trachea/drug effects , Trachea/physiology
11.
J Biol Chem ; 276(28): 25647-50, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11384963

ABSTRACT

BRCA1 gene is a tumor suppressor for breast and ovarian cancers with the putative role in DNA repair and transcription. To characterize the role of BRCA1 in transcriptional regulation, we analyzed gene expression profiles of mouse embryonic stem cells deficient in BRCA1 using microarray technology. We found that loss of BRCA1 correlated with decreased expression of several groups of genes including stress response genes, cytoskeleton genes, and genes involved in protein synthesis and degradation. Previous study showed that BRCA1 is a transcriptional co-activator of p53 protein; however the majority of p53 target genes remained at the same expression levels in BRCA1 knockout cells as in the wild type cells. The only p53 target gene down-regulated with the loss of BRCA1 was 14-3-3 sigma, a major G(2)/M checkpoint control gene. Similar to cells with decreased 14-3-3 sigma activity, BRCA1-deficient cells were unable to sustain G(2)/M growth arrest after exposure to ionizing radiation. We find that BRCA1 induction of 14-3-3 sigma requires the presence of wild type p53 and can be regulated by a minimal p53 response element.


Subject(s)
Biomarkers, Tumor , Exonucleases , Genes, BRCA1 , Neoplasm Proteins , Proteins/genetics , Stem Cells/physiology , 14-3-3 Proteins , Animals , Exoribonucleases , Gene Expression Regulation, Developmental/physiology , Genes, p53 , Mice , Transcription, Genetic/physiology
12.
Hear Res ; 156(1-2): 17-30, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11377879

ABSTRACT

The transduction of the auditory signal is dependent on the flow of ions within the inner ear. We have generated mice deficient in NKCC1, an ion cotransporter that is thought to be involved in the secretion of K+ by the strial marginal cells. Inner ear histology revealed partial to almost total absence of the scala media and collapse of Reissner's membrane. Ultrastructural analysis showed that Reissner's membrane consists of 3-4 cell layers instead of the usual two, and a substance of unknown composition is present between Reissner's membrane and underlying structures. Within the tunnel of Corti, hair cells and supporting cells were difficult to identify. The location of the tectorial membrane was altered, and a precipitate was observed surrounding it. Severe structural defects were noted in the interdental cells and Boettcher cells, and mild defects were observed in the stria vascularis and in type II and type IV fibrocytes. The finding that major defects occur predominantly in cells that are not known to express NKCC1 suggests that loss of NKCC1 results in functional defects in cells expressing NKCC1 and a morphological effect on cell populations downstream in the proposed K+ recycling pathway.


Subject(s)
Carrier Proteins/physiology , Ear, Inner/ultrastructure , Animals , Carrier Proteins/genetics , Cell Line , Ear, Inner/pathology , Mice , Mice, Knockout/genetics , Microscopy, Electron , Sodium-Potassium-Chloride Symporters , Stria Vascularis/pathology , Stria Vascularis/ultrastructure , Tectorial Membrane/pathology , Tectorial Membrane/ultrastructure
13.
Proc Natl Acad Sci U S A ; 98(6): 3369-74, 2001 Mar 13.
Article in English | MEDLINE | ID: mdl-11248085

ABSTRACT

The pregnane X receptor (PXR) is the molecular target for catatoxic steroids such as pregnenolone 16alpha-carbonitrile (PCN), which induce cytochrome P450 3A (CYP3A) expression and protect the body from harmful chemicals. In this study, we demonstrate that PXR is activated by the toxic bile acid lithocholic acid (LCA) and its 3-keto metabolite. Furthermore, we show that PXR regulates the expression of genes involved in the biosynthesis, transport, and metabolism of bile acids including cholesterol 7alpha-hydroxylase (Cyp7a1) and the Na(+)-independent organic anion transporter 2 (Oatp2). Finally, we demonstrate that activation of PXR protects against severe liver damage induced by LCA. Based on these data, we propose that PXR serves as a physiological sensor of LCA, and coordinately regulates gene expression to reduce the concentrations of this toxic bile acid. These findings suggest that PXR agonists may prove useful in the treatment of human cholestatic liver disease.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cholestasis, Intrahepatic/metabolism , Lithocholic Acid/metabolism , Liver/injuries , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , Animals , Cholestasis, Intrahepatic/prevention & control , Cholesterol 7-alpha-Hydroxylase/genetics , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Female , Gene Expression Regulation, Enzymologic , Lithocholic Acid/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases, N-Demethylating/genetics , Pregnane X Receptor , Pregnenolone Carbonitrile/pharmacology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism
14.
J Clin Invest ; 107(5): 603-10, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238561

ABSTRACT

The importance of arachidonic acid metabolites (termed eicosanoids), particularly those derived from the COX-1 and COX-2 pathways (termed prostanoids), in platelet homeostasis has long been recognized. Thromboxane is a potent agonist, whereas prostacyclin is an inhibitor of platelet aggregation. In contrast, the effect of prostaglandin E2 (PGE2) on platelet aggregation varies significantly depending on its concentration. Low concentrations of PGE2 enhance platelet aggregation, whereas high PGE2 levels inhibit aggregation. The mechanism for this dual action of PGE2 is not clear. This study shows that among the four PGE2 receptors (EP1-EP4), activation of EP3 is sufficient to mediate the proaggregatory actions of low PGE2 concentration. In contrast, the prostacyclin receptor (IP) mediates the inhibitory effect of higher PGE2 concentrations. Furthermore, the relative activation of these two receptors, EP3 and IP, regulates the intracellular level of cAMP and in this way conditions the response of the platelet to aggregating agents. Consistent with these findings, loss of the EP3 receptor in a model of venous inflammation protects against formation of intravascular clots. Our results suggest that local production of PGE2 during an inflammatory process can modulate ensuing platelet responses.


Subject(s)
Cyclic AMP/biosynthesis , Dinoprostone/pharmacology , Platelet Aggregation , Receptors, Prostaglandin E/metabolism , Animals , Calcium/metabolism , Female , Male , Mice , Mice, Knockout , Models, Biological , Platelet Aggregation/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Epoprostenol , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP3 Subtype , Venous Thrombosis/pathology
15.
Proc Natl Acad Sci U S A ; 98(3): 1059-64, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11158594

ABSTRACT

The transition to pulmonary respiration following birth requires rapid alterations in the structure of the mammalian cardiovascular system. One dramatic change that occurs is the closure and remodeling of the ductus arteriosus (DA), an arterial connection in the fetus that directs blood flow away from the pulmonary circulation. A role for prostaglandins in regulating the closure of this vessel has been supported by pharmacological and genetic studies. The production of prostaglandins is dependent on two cyclooxygenases (COX-1 and COX-2), which are encoded by separate genes. We report here that the absence of either or both COX isoforms in mice does not result in premature closure of the DA in utero. However, 35% of COX-2(-/-) mice die with a patent DA within 48 h of birth. In contrast, the absence of only the COX-1 isoform does not affect closure of the DA. The mortality (35%) and patent DA incidence due to absence of COX-2 is, however, significantly increased (79%) when one copy of the gene encoding COX-1 is also inactivated. Furthermore, 100% of the mice deficient in both isoforms die with a patent DA within 12 h of birth, indicating that in COX-2-deficient mice, the contribution of COX-1 to DA closure is gene dosage-dependent. Together, these data establish roles for COX-1, and especially for COX-2, in the transition of the cardiopulmonary circulation at birth.


Subject(s)
Ductus Arteriosus, Patent/genetics , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Animals, Newborn , Cyclooxygenase 1 , Cyclooxygenase 2 , Death , Ductus Arteriosus/pathology , Ductus Arteriosus, Patent/epidemiology , Female , Genomic Imprinting , Genotype , Isoenzymes/deficiency , Isoenzymes/genetics , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Prostaglandin-Endoperoxide Synthases/deficiency , Prostaglandin-Endoperoxide Synthases/genetics , Time Factors
16.
J Biol Chem ; 276(1): 125-32, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11016935

ABSTRACT

The P2X(7) receptor (P2X(7)R) is an ATP-gated ion channel expressed by monocytes and macrophages. To directly address the role of this receptor in interleukin (IL)-1 beta post-translational processing, we have generated a P2X(7)R-deficient mouse line. P2X(7)R(-/-) macrophages respond to lipopolysaccharide and produce levels of cyclooxygenase-2 and pro-IL-1 beta comparable with those generated by wild-type cells. In response to ATP, however, pro-IL-1 beta produced by the P2X(7)R(-/-) cells is not externalized or activated by caspase-1. Nigericin, an alternate secretion stimulus, promotes release of 17-kDa IL-1 beta from P2X(7)R(-/-) macrophages. In response to in vivo lipopolysaccharide injection, both wild-type and P2X(7)R(-/-) animals display increases in peritoneal lavage IL-6 levels but no detectable IL-1. Subsequent ATP injection to wild-type animals promotes an increase in IL-1, which in turn leads to additional IL-6 production; similar increases did not occur in ATP-treated, LPS-primed P2X(7)R(-/-) animals. Absence of the P2X(7)R thus leads to an inability of peritoneal macrophages to release IL-1 in response to ATP. As a result of the IL-1 deficiency, in vivo cytokine signaling cascades are impaired in P2X(7)R-deficient animals. Together these results demonstrate that P2X(7)R activation can provide a signal that leads to maturation and release of IL-1 beta and initiation of a cytokine cascade.


Subject(s)
Gene Deletion , Interleukin-1/biosynthesis , Protein Precursors/biosynthesis , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/physiology , Adenosine Triphosphate/pharmacology , Animals , Cells, Cultured , Cyclooxygenase 2 , Enzyme Induction/drug effects , Fluorescent Dyes/metabolism , Gene Targeting , Inflammation/genetics , Interleukin-1/metabolism , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Isoenzymes/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Nigericin/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational/drug effects , Purinergic P2 Receptor Agonists , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7
17.
Am J Physiol Heart Circ Physiol ; 280(1): H327-33, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11123248

ABSTRACT

The kidney plays a central role in long-term regulation of arterial blood pressure and salt and water homeostasis. This is achieved in part by the local actions of paracrine and autacoid mediators such as the arachidonic acid-prostanoid system. The present study tested the role of specific PGE(2) E-prostanoid (EP) receptors in the regulation of renal hemodynamics and vascular reactivity to PGE(2). Specifically, we determined the extent to which the EP(2) and EP(3) receptor subtypes mediate the actions of PGE(2) on renal vascular tone. Renal blood flow (RBF) was measured by ultrasonic flowmetry, whereas vasoactive agents were injected directly into the renal artery of male mice. Studies were performed on two independent mouse lines lacking either EP(2) or EP(3) (-/-) receptors and the results were compared with wild-type controls (+/+). Our results do not support a unique role of the EP(2) receptor in regulating overall renal hemodynamics. Baseline renal hemodynamics in EP(2)-/- mice [RBF EP(2)-/-: 5.3 +/- 0.8 ml. min(-1). 100 g kidney wt(-1); renal vascular resistance (RVR) 19.7 +/- 3.6 mmHg. ml(-1). min. g kidney wt] did not differ statistically from control mice (RBF +/+: 4.0 +/- 0.5 ml. min(-1). 100 g kidney wt(-1); RVR +/+: 25.4 +/- 4.9 mmHg. ml(-1). min. 100 g kidney wt(-1)). This was also the case for the peak RBF increase after local PGE(2) (500 ng) injection into the renal artery (EP(2)-/-: 116 +/- 4 vs. +/+: 112 +/- 2% baseline RBF). In contrast, we found that the absence of EP(3) receptors in EP(3)-/- mice caused a significant increase (43%) in basal RBF (7.9 +/- 0.8 ml. min(-1). g kidney wt(-1), P < 0.05 vs. +/+) and a significant decrease (41%) in resting RVR (11.6 +/- 1.4 mmHg. ml(-1). min. g kidney wt(-1), P < 0.05 vs. +/+). Local administration of 500 ng of PGE(2) into the renal artery caused more pronounced renal vasodilation in EP(3)-/- mice (128 +/- 2% of basal RBF, P < 0.05 vs. +/+). We conclude that EP(3 )receptors mediate vasoconstriction in the kidney of male mice and its actions are tonically active in the basal state. Furthermore, EP(3) receptors are capable of buffering PGE(2)-mediated renal vasodilation.


Subject(s)
Dinoprostone/pharmacology , Receptors, Prostaglandin E/physiology , Renal Circulation/physiology , Animals , Dinoprostone/metabolism , Genotype , Kidney/blood supply , Kidney/diagnostic imaging , Male , Mice , Mice, Knockout , Microcirculation , Mutation , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP3 Subtype , Renal Circulation/drug effects , Ultrasonography
18.
Am J Physiol Gastrointest Liver Physiol ; 279(4): G707-18, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11005757

ABSTRACT

The Na(+)-K(+)-2Cl(-) cotransporter (NKCC1) located on the basolateral membrane of intestinal epithelia has been postulated to be the major basolateral Cl(-) entry pathway. With targeted mutagenesis, mice deficient in the NKCC1 protein were generated. The basal short-circuit current did not differ between normal and NKCC1 -/- jejuna. In the -/- jejuna, the forskolin response (22 microA/cm(2); bumetanide insensitive) was significantly attenuated compared with the bumetanide-sensitive response (52 microA/cm(2)) in normal tissue. Ion-replacement studies demonstrated that the forskolin response in the NKCC1 -/- jejuna was HCO(3)(-) dependent, whereas in the normal jejuna it was independent of the HCO(3)(-) concentration in the buffer. NKCC1 -/- ceca exhibited a forskolin response that did not differ significantly from that of normal ceca, but unlike that of normal ceca, was bumetanide insensitive. Ion-substitution studies suggested that basolateral HCO(3)(-) as well as Cl(-) entry (via non-NKCC1) paths played a role in the NKCC1 -/- secretory response. In contrast to cystic fibrosis mice, which lack both basal and stimulated Cl(-) secretion and exhibit severe intestinal pathology, the absence of intestinal pathology in NKCC1 -/- mice likely reflects the ability of the intestine to secrete HCO(3)(-) and Cl(-) by basolateral entry mechanisms independent of NKCC1.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Cecum/physiology , Chlorides/metabolism , Intestinal Mucosa/physiology , Jejunum/physiology , Potassium/metabolism , Sodium/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Bicarbonates/metabolism , Bumetanide/pharmacology , Cecum/drug effects , Colforsin/pharmacology , Glucose/pharmacology , Intestinal Mucosa/drug effects , Jejunum/drug effects , Kinetics , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Knockout , Models, Biological , Mutagenesis , Sodium-Potassium-Chloride Symporters
19.
Cancer Res ; 60(13): 3461-9, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10910057

ABSTRACT

Women who inherit mutations in the breast cancer susceptibility genes, BRCA1 and BRCA2, are predisposed to the development of breast and ovarian cancer. We used mice with a Brca1 mutation on a BALB/cJ inbred background (BALB/cB1+/- mice) or a Brca2 genetic alteration on the 129/SvEv genetic background (129B2+/- mice) to investigate potential gene-environment interactions between defects in these genes and treatment with the highly estrogenic compound diethylstilbestrol (DES). Beginning at 3 weeks of age, BALB/cB1+/-, 129B2+/-, and wild-type female mice were fed a control diet or a diet containing 640 ppb DES for 26 weeks. DES treatment caused vaginal epithelial hyperplasia and hyperkeratosis, uterine inflammation, adenomyosis, and fibrosis, as well as oviductal smooth muscle hypertrophy. The severity of the DES response was mouse strain specific. The estrogen-responsive 129/SvEv strain exhibited an extreme response in the reproductive tract, whereas the effect in BALB/cJ and C3H/HeN(MMTV-) mice was less severe. The Brca1 and Brca2 genetic alterations influenced the phenotypic response of BALB/cJ and 129/SvEv inbred strains, respectively, to DES in the mammary gland and ovary. The mammary duct branching morphology was inhibited in DES-treated BALB/cB1+/- mice compared with similarly treated BALB/cB1+/+ littermates. In addition, the majority of BALB/cB1+/- mice had atrophied ovaries, whereas wild-type littermates were largely diagnosed with arrested follicular development. The mammary ductal architecture in untreated 129B2+/- mice revealed a subtle inhibited branching phenotype that was enhanced with DES treatment. However, no significant differences were observed in ovarian pathology between 129B2+/+ and 129B2+/- mice. These data suggest that estrogenic compounds may modulate mammary gland or ovarian morphology in BALB/cB1+/- and 129B2+/- mice. These observations are consistent with the hypothesis that compromised DNA repair processes in cells harboring Brca1 or Brca2 mutations lead to inhibited growth and differentiation compared with the proliferative response of wild-type cells to DES treatment.


Subject(s)
Diethylstilbestrol/toxicity , Genes, BRCA1/genetics , Mammary Glands, Animal/pathology , Neoplasm Proteins/genetics , Ovary/pathology , Transcription Factors/genetics , Animals , BRCA2 Protein , Carcinogens/toxicity , Chimera , Crosses, Genetic , Endometriosis/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Fallopian Tubes/drug effects , Fallopian Tubes/pathology , Female , Fibrosis/chemically induced , Genetic Markers , Heterozygote , Hypertrophy , Inflammation , Mammary Glands, Animal/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Transgenic , Muscle, Smooth/drug effects , Muscle, Smooth/pathology , Ovary/drug effects , Phenotype , Uterus/drug effects , Uterus/pathology , Vagina/drug effects , Vagina/pathology
20.
Circulation ; 101(24): 2833-40, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10859290

ABSTRACT

BACKGROUND: Isoprostanes (iPs) are free radical-catalyzed products of arachidonic acid that reflect lipid peroxidation in vivo. Several iPs exert biological effects in vitro and may contribute to the functional consequences of oxidant stress. For example, iPF(2alpha)-III (8-iso PGF(2alpha)) and iPE(2)-III modulate platelet function and vascular tone. Although these effects are blocked by antagonists of the receptor (TP) for the cyclooxygenase product thromboxane A(2), it has been speculated that the iPs may activate a receptor related to, but distinct from, the TP. METHODS AND RESULTS: Transgenic mice (TPOEs) were generated in which the TP-beta isoform was under the control of the preproendothelin promoter. They overexpressed TP-beta in the vasculature but not in platelets and exhibited an exaggerated pressor response to infused iPF(2alpha)-III compared with wild-type mice. This was blocked by TP antagonism. The platelet response to the iP was unaltered in TPOEs compared with wild-type mice. By contrast, both the pressor response to iPF(2alpha)-III and its effects on platelet function were abolished in mice lacking the TP gene. This was also true of the effects of infused iPE(2)-III on mean arterial pressure and platelet aggregation. CONCLUSIONS: Both iPF(2alpha)-III and iPE(2)-III exert their effects on platelet function and vascular tone in vivo by acting as incidental ligands at membrane TPs rather than via a distinct iP receptor. Activation of TPs by iPs may be of importance in syndromes in which cyclooxygenase activation and oxidant stress coincide, such as in atherosclerosis and reperfusion after tissue ischemia.


Subject(s)
Cardiovascular System/drug effects , Dinoprost/analogs & derivatives , Dinoprost/pharmacology , Receptors, Thromboxane/physiology , Angiotensin II/pharmacology , Animals , Blood Platelets/drug effects , Blood Platelets/physiology , Blood Pressure/drug effects , Mice , Mice, Transgenic/genetics , Platelet Aggregation/drug effects , Protein Isoforms/genetics , Receptors, Thromboxane/genetics , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...