Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
2.
Adv Exp Med Biol ; 1274: 101-135, 2020.
Article in English | MEDLINE | ID: mdl-32894509

ABSTRACT

Intensive research in the field of sphingolipids has revealed diverse roles in cell biological responses and human health and disease. This immense molecular family is primarily represented by the bioactive molecules ceramide, sphingosine, and sphingosine 1-phosphate (S1P). The flux of sphingolipid metabolism at both the subcellular and extracellular levels provides multiple opportunities for pharmacological intervention. The caveat is that perturbation of any single node of this highly regulated flux may have effects that propagate throughout the metabolic network in a dramatic and sometimes unexpected manner. Beginning with S1P, the receptors for which have thus far been the most clinically tractable pharmacological targets, this review will describe recent advances in therapeutic modulators targeting sphingolipids, their chaperones, transporters, and metabolic enzymes.


Subject(s)
Metabolic Networks and Pathways/drug effects , Models, Biological , Molecular Targeted Therapy , Sphingolipids/metabolism , Ceramides/metabolism , Humans , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism
4.
FASEB J ; 34(2): 2840-2852, 2020 02.
Article in English | MEDLINE | ID: mdl-31908031

ABSTRACT

Eicosanoids are powerful mediators of inflammation and are known to drive both the progression and regression of arthritis. We previously reported the infection of C3H 5-lipoxygenase (LO)-deficient mice with Borrelia burgdorferi results in prolonged nonresolving Lyme arthritis. Here we define the role of the 5-LO metabolite leukotriene (LT)B4 and its high-affinity receptor, BLT1, in this response. C3H and C3H BLT1-/- mice were infected with B. burgdorferi and arthritis progression was monitored by ankle swelling over time. Similar to 5-LO-/- mice, BLT1-/- mice developed nonresolving Lyme arthritis characterized by increased neutrophils in the joint at later time points than WT mice, but with fewer apoptotic (caspase-3+ ) neutrophils. In vitro, BLT1-/- neutrophils were defective in their ability to undergo apoptosis due to the lack of LTB4 -mediated down-regulation of cAMP, subsequent failure to induce Death-Inducing Signaling Complex (DISC) components, and decreased FasL and CD36 expression. Inhibition of adenylyl cyclase with SQ 22,536 restored BLT1-/- BMN apoptosis, FasL and CD36 expression, and clearance by macrophages. We conclude that LTB4/BLT1 signaling has an unexpected critical role in mediating neutrophil apoptosis via the down-regulation of cAMP. Loss of BLT1 signaling led to defective clearance of neutrophils from the inflamed joint and failed arthritis resolution.


Subject(s)
Apoptosis , Borrelia burgdorferi/metabolism , Lyme Disease/metabolism , Neutrophils/metabolism , Receptors, Leukotriene B4/metabolism , Signal Transduction , Animals , Disease Models, Animal , Lyme Disease/genetics , Lyme Disease/pathology , Mice , Mice, Knockout , Neutrophils/pathology , Receptors, Leukotriene B4/genetics
5.
Sci Adv ; 5(10): eaax2011, 2019 10.
Article in English | MEDLINE | ID: mdl-31633020

ABSTRACT

Posthemorrhagic hydrocephalus (PHH) in premature infants is a common neurological disorder treated with invasive neurosurgical interventions. Patients with PHH lack effective therapeutic interventions and suffer chronic comorbidities. Here, we report a murine lysophosphatidic acid (LPA)-induced postnatal PHH model that maps neurodevelopmentally to premature infants, a clinically accessible high-risk population, and demonstrates ventriculomegaly with increased intracranial pressure. Administration of LPA, a blood-borne signaling lipid, acutely disrupted the ependymal cells that generate CSF flow, which was followed by cell death, phagocytosis, and ventricular surface denudation. This mechanism is distinct from a previously reported fetal model that induces PHH through developmental alterations. Analyses of LPA receptor-null mice identified LPA1 and LPA3 as key mediators of PHH. Pharmacological blockade of LPA1 prevented PHH in LPA-injected animals, supporting the medical tractability of LPA receptor antagonists in preventing PHH and negative CNS sequelae in premature infants.


Subject(s)
Infant, Premature, Diseases/pathology , Receptors, Lysophosphatidic Acid/metabolism , Animals , Animals, Newborn , Apoptosis , Brain/metabolism , Brain/pathology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Ependyma/cytology , Ependyma/metabolism , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Infant, Premature, Diseases/chemically induced , Infant, Premature, Diseases/prevention & control , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Lysophospholipids/toxicity , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Phagocytosis , Propionates/pharmacology , Propionates/therapeutic use , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics
6.
J Lipid Res ; 60(11): 1912-1921, 2019 11.
Article in English | MEDLINE | ID: mdl-31462513

ABSTRACT

HDL-bound ApoM and albumin are protein chaperones for the circulating bioactive lipid, sphingosine 1-phosphate (S1P); in this role, they support essential extracellular S1P signaling functions in the vascular and immune systems. We previously showed that ApoM- and albumin-bound S1P exhibit differences in receptor activation and biological functions. Whether the physiological functions of S1P require chaperones is not clear. We examined ApoM-deficient, albumin-deficient, and double-KO (DKO) mice for circulatory S1P and its biological functions. In albumin-deficient mice, ApoM was upregulated, thus enabling S1P functions in embryonic development and postnatal adult life. The Apom:Alb DKO mice reproduced, were viable, and exhibited largely normal vascular and immune functions, which suggested sufficient extracellular S1P signaling. However, Apom:Alb DKO mice had reduced levels (∼25%) of plasma S1P, suggesting that novel S1P chaperones exist to mediate S1P functions. In this study, we report the identification of ApoA4 as a novel S1P binding protein. Recombinant ApoA4 bound to S1P, activated multiple S1P receptors, and promoted vascular endothelial barrier function, all reflective of its function as a S1P chaperone in the absence of ApoM and albumin. We suggest that multiple S1P chaperones evolved to support complex and essential extracellular signaling functions of this lysolipid mediator in a redundant manner.


Subject(s)
Apolipoproteins A/metabolism , Apolipoproteins M/deficiency , Lysophospholipids/metabolism , Serum Albumin/deficiency , Sphingosine/analogs & derivatives , Amino Acid Sequence , Animals , Apolipoproteins A/chemistry , Apolipoproteins M/genetics , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/metabolism
7.
Front Immunol ; 10: 404, 2019.
Article in English | MEDLINE | ID: mdl-30941120

ABSTRACT

Background: Although myelin is composed of mostly lipids, the pathological role of myelin lipids in demyelinating diseases remains elusive. The principal lipid of the myelin sheath is ß-galactosylceramide (ß-Galcer). Its α-anomer (α-Galcer) has been demonstrated to be antigenically presented by macrophages via CD1d, a MHC class I-like molecule. Myelin, which is mostly composed of ß-Galcer, has been long considered as an immunologically-inert neuron insulator, because the antigen-binding cleft of CD1d is highly α-form-restricted. Results: Here, we report that CD1d-mediated antigenic presentation of myelin-derived galactosylceramide (Mye-GalCer) by macrophages contributed significantly to the progression of experimental autoimmune encephalomyelitis (EAE). Surprisingly, this presentation was recognizable by α-Galcer:CD1d-specific antibody (clone L363), but incapable of triggering expansion of iNKT cells and production of iNKT signature cytokines (IFNγ and IL-4). Likewise, a synthesized analog of Mye-Galcer, fluorinated α-C-GalCer (AA2), while being efficiently presented via CD1d on macrophages, failed to stimulate production of IFNγ and IL-4. However, AA2 significantly exacerbated EAE progression. Further analyses revealed that the antigenic presentations of both Mye-GalCer and its analog (AA2) in α-form via CD1d promoted IL-17 production from T cells, leading to elevated levels of IL-17 in EAE spinal cords and sera. The IL-17 neutralizing antibody significantly reduced the severity of EAE symptoms in AA2-treated mice. Furthermore, D-sphingosine, a lipid possessing the same hydrophobic base as ceramide but without a carbohydrate residue, efficiently blocked this glycolipid antigen presentation both in vitro and in spinal cords of EAE mice, and significantly decreased IL-17 and ameliorated the pathological symptoms. Conclusion: Our findings reveal a novel pathway from the presentation of Mye-GalCer to IL-17 production, and highlight the promising therapeutic potential of D-sphingosine for the human disorder of multiple sclerosis.


Subject(s)
Antigen Presentation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Galactosylceramides/immunology , Macrophages/immunology , Myelin Sheath/immunology , Sphingosine/immunology , Animals , Antigen Presentation/drug effects , Autoantigens/chemistry , Autoantigens/immunology , Female , Glycolipids/immunology , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Myelin Sheath/chemistry , Sphingosine/pharmacology
8.
Annu Rev Pharmacol Toxicol ; 59: 149-170, 2019 01 06.
Article in English | MEDLINE | ID: mdl-30625282

ABSTRACT

Fingolimod (FTY720, Gilenya) was the first US Food and Drug Administration-approved oral therapy for relapsing forms of multiple sclerosis (MS). Research on modified fungal metabolites converged with basic science studies that had identified lysophospholipid (LP) sphingosine 1-phosphate (S1P) receptors, providing mechanistic insights on fingolimod while validating LP receptors as drug targets. Mechanism of action (MOA) studies identified receptor-mediated processes involving the immune system and the central nervous system (CNS). These dual actions represent a more general theme for S1P and likely other LP receptor modulators. Fingolimod's direct CNS activities likely contribute to its efficacy in MS, with particular relevance to treating progressive disease stages and forms that involve neurodegeneration. The evolving understanding of fingolimod's MOA has provided strategies for developing next-generation compounds with superior attributes, suggesting new ways to target S1P as well as other LP receptor modulators for novel therapeutics in the CNS and other organ systems.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Multiple Sclerosis/drug therapy , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Humans , Lysophospholipids/metabolism , Multiple Sclerosis/metabolism , Sphingosine-1-Phosphate Receptors/metabolism
9.
Trends Pharmacol Sci ; 39(11): 953-966, 2018 11.
Article in English | MEDLINE | ID: mdl-30343728

ABSTRACT

Lysophospholipids (LPLs), particularly sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA), are bioactive lipid modulators of cellular homeostasis and pathology. The discovery and characterization of five S1P- and six LPA-specific G protein-coupled receptors (GPCRs), S1P1-5 and LPA1-6, have expanded their known involvement in all mammalian physiological systems. Resolution of the S1P1, LPA1, and LPA6 crystal structures has fueled the growing interest in these receptors and their ligands as targets for pharmacological manipulation. In this review, we have attempted to provide an integrated overview of the three crystallized LPL GPCRs with biochemical and physiological structure-function data. Finally, we provide a novel discussion of how chaperones for LPLs may be considered when extrapolating crystallographic and computational data toward understanding actual biological interactions and phenotypes.


Subject(s)
Receptors, Lysophospholipid/chemistry , Animals , Humans , Ligands , Lysophospholipids/metabolism , Protein Conformation , Receptors, Lysophospholipid/metabolism
10.
Sci Signal ; 8(389): ra79, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26268607

ABSTRACT

The sphingosine 1-phosphate receptor 1 (S1P1) is abundant in endothelial cells, where it regulates vascular development and microvascular barrier function. In investigating the role of endothelial cell S1P1 in adult mice, we found that the endothelial S1P1 signal was enhanced in regions of the arterial vasculature experiencing inflammation. The abundance of proinflammatory adhesion proteins, such as ICAM-1, was enhanced in mice with endothelial cell-specific deletion of S1pr1 and suppressed in mice with endothelial cell-specific overexpression of S1pr1, suggesting a protective function of S1P1 in vascular disease. The chaperones ApoM(+)HDL (HDL) or albumin bind to sphingosine 1-phosphate (S1P) in the circulation; therefore, we tested the effects of S1P bound to each chaperone on S1P1 signaling in cultured human umbilical vein endothelial cells (HUVECs). Exposure of HUVECs to ApoM(+)HDL-S1P, but not to albumin-S1P, promoted the formation of a cell surface S1P1-ß-arrestin 2 complex and attenuated the ability of the proinflammatory cytokine TNFα to activate NF-κB and increase ICAM-1 abundance. Although S1P bound to either chaperone induced MAPK activation, albumin-S1P triggered greater Gi activation and receptor endocytosis. Endothelial cell-specific deletion of S1pr1 in the hypercholesterolemic Apoe(-/-) mouse model of atherosclerosis enhanced atherosclerotic lesion formation in the descending aorta. We propose that the ability of ApoM(+)HDL to act as a biased agonist on S1P1 inhibits vascular inflammation, which may partially explain the cardiovascular protective functions of HDL.


Subject(s)
Atherosclerosis/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Lysophospholipids/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Vasculitis/metabolism , Animals , Apolipoproteins/genetics , Apolipoproteins/metabolism , Apolipoproteins M , Atherosclerosis/genetics , Atherosclerosis/pathology , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/pathology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lipocalins/genetics , Lipocalins/metabolism , Lipoproteins, HDL/genetics , Lysophospholipids/genetics , Mice , Mice, Knockout , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/genetics , Sphingosine/genetics , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , Vasculitis/genetics , Vasculitis/pathology
11.
Nature ; 523(7560): 342-6, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26053123

ABSTRACT

Lipid mediators influence immunity in myriad ways. For example, circulating sphingosine-1-phosphate (S1P) is a key regulator of lymphocyte egress. Although the majority of plasma S1P is bound to apolipoprotein M (ApoM) in the high-density lipoprotein (HDL) particle, the immunological functions of the ApoM-S1P complex are unknown. Here we show that ApoM-S1P is dispensable for lymphocyte trafficking yet restrains lymphopoiesis by activating the S1P1 receptor on bone marrow lymphocyte progenitors. Mice that lacked ApoM (Apom(-/-)) had increased proliferation of Lin(-) Sca-1(+) cKit(+) haematopoietic progenitor cells (LSKs) and common lymphoid progenitors (CLPs) in bone marrow. Pharmacological activation or genetic overexpression of S1P1 suppressed LSK and CLP cell proliferation in vivo. ApoM was stably associated with bone marrow CLPs, which showed active S1P1 signalling in vivo. Moreover, ApoM-bound S1P, but not albumin-bound S1P, inhibited lymphopoiesis in vitro. Upon immune stimulation, Apom(-/-) mice developed more severe experimental autoimmune encephalomyelitis, characterized by increased lymphocytes in the central nervous system and breakdown of the blood-brain barrier. Thus, the ApoM-S1P-S1P1 signalling axis restrains the lymphocyte compartment and, subsequently, adaptive immune responses. Unique biological functions imparted by specific S1P chaperones could be exploited for novel therapeutic opportunities.


Subject(s)
Apolipoproteins/metabolism , Central Nervous System/pathology , Lipoproteins, HDL/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Lymphopoiesis , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Apolipoproteins/deficiency , Apolipoproteins/genetics , Apolipoproteins M , Blood-Brain Barrier/pathology , Cell Movement , Cell Proliferation/genetics , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Fingolimod Hydrochloride/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lymphocytes/immunology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lysophospholipids/agonists , Lysophospholipids/blood , Lysophospholipids/genetics , Male , Mice , Mice, Inbred C57BL , Protein Binding , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/agonists , Sphingosine/blood , Sphingosine/genetics , Sphingosine/metabolism
12.
Immunology ; 142(3): 347-53, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24597601

ABSTRACT

Sphingosine-1-phosphate (S1P) is a lipid second messenger that signals via five G protein-coupled receptors (S1P1-5 ). S1P receptor (S1PR) signalling is associated with a wide variety of physiological processes including lymphocyte biology, their recirculation and determination of T-cell phenotypes. The effect of FTY720 (Fingolimod, Gilenya™) to regulate lymphocyte egress and to ameliorate paralysis in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis led to the use of FTY720 as a first-line oral agent for treatment of relapsing-remitting multiple sclerosis. However, a significant body of research suggests that S1P signalling may participate in diverse immune regulatory functions other than lymphocyte trafficking. This review article discusses the current knowledge of S1P signalling in the fate and function of T regulatory, T helper type 17 and memory T cells in health and disease.


Subject(s)
Receptors, Lysosphingolipid/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Humans , Sphingosine-1-Phosphate Receptors , T-Lymphocytes/cytology
13.
J Lipid Res ; 55(8): 1596-608, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24459205

ABSTRACT

Sphingosine 1-phosphate (S1P) is a membrane-derived lysophospholipid that acts primarily as an ex-tracellular signaling molecule. Signals initiated by S1P are transduced by five G protein-coupled receptors, named S1P1-5 Cellular and temporal expression of the S1P receptors (S1PRs) determine their specific roles in various organ systems, but they are particularly critical for regulation of the cardiovascular, immune, and nervous systems, with the most well-known contributions of S1PR signaling being modulation of vascular barrier function, vascular tone, and regulation of lymphocyte trafficking. However, our knowledge of S1PR biology is rapidly increasing as they become attractive therapeutic targets in several diseases, such as chronic inflammatory pathologies, autoimmunity, and cancer. Understanding how the S1PRs regulate interactions between biological systems will allow for greater efficacy in this novel therapeutic strategy as well as characterization of complex physiological networks. Because of the rapidly expanding body of research, this review will focus on the most recent advances in S1PRs.


Subject(s)
Autoimmune Diseases/metabolism , Lysophospholipids/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Humans , Sphingosine/metabolism
14.
Nat Immunol ; 14(11): 1166-72, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24076635

ABSTRACT

Sphingosine 1-phosphate (S1P) signaling regulates lymphocyte egress from lymphoid organs into systemic circulation. The sphingosine phosphate receptor 1 (S1P1) agonist FTY-720 (Gilenya) arrests immune trafficking and prevents multiple sclerosis (MS) relapses. However, alternative mechanisms of S1P-S1P1 signaling have been reported. Phosphoproteomic analysis of MS brain lesions revealed S1P1 phosphorylation on S351, a residue crucial for receptor internalization. Mutant mice harboring an S1pr1 gene encoding phosphorylation-deficient receptors (S1P1(S5A)) developed severe experimental autoimmune encephalomyelitis (EAE) due to autoimmunity mediated by interleukin 17 (IL-17)-producing helper T cells (TH17 cells) in the peripheral immune and nervous system. S1P1 directly activated the Jak-STAT3 signal-transduction pathway via IL-6. Impaired S1P1 phosphorylation enhances TH17 polarization and exacerbates autoimmune neuroinflammation. These mechanisms may be pathogenic in MS.


Subject(s)
Brain/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Interleukin-17/metabolism , Lysophospholipids/metabolism , Multiple Sclerosis/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction/immunology , Sphingosine/analogs & derivatives , Animals , Autopsy , Brain/immunology , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation , Humans , Inflammation , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Janus Kinases/genetics , Janus Kinases/immunology , Janus Kinases/metabolism , Lysophospholipids/immunology , Mice , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Phosphorylation , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Sphingosine/immunology , Sphingosine/metabolism , Th17 Cells
15.
J Nutr ; 142(8): 1582-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22695969

ABSTRACT

Dietary ingestion of (n-3) PUFA alters the production of eicosanoids and can suppress chronic inflammatory and autoimmune diseases. The extent of changes in eicosanoid production during an infection of mice fed a diet high in (n-3) PUFA, however, has not, to our knowledge, been reported. We fed mice a diet containing either 18% by weight soybean oil (SO) or a mixture with fish oil (FO), FO:SO (4:1 ratio), for 2 wk and then infected them with Borrelia burgdorferi. We used an MS-based lipidomics approach and quantified changes in eicosanoid production during Lyme arthritis development over 21 d. B. burgdorferi infection induced a robust production of prostanoids, mono-hydroxylated metabolites, and epoxide-containing metabolites, with 103 eicosanoids detected of the 139 monitored. In addition to temporal and compositional changes in the eicosanoid profile, dietary FO substitution increased the accumulation of 15-deoxy PGJ(2), an antiinflammatory metabolite derived from arachidonic acid. Chiral analysis of the mono-hydroxylated metabolites revealed they were generated from primarily nonenzymatic mechanisms. Although dietary FO substitution reduced the production of inflammatory (n-6) fatty acid-derived eicosanoids, no change in the host inflammatory response or development of disease was detected.


Subject(s)
Dietary Fats, Unsaturated/pharmacology , Eicosanoids/metabolism , Fish Oils/pharmacology , Joints/metabolism , Lyme Disease/diet therapy , Lyme Disease/metabolism , Animal Feed , Animals , Dietary Fats, Unsaturated/administration & dosage , Fatty Acids/blood , Fatty Acids/chemistry , Fatty Acids/metabolism , Female , Fish Oils/administration & dosage , Hindlimb , Hot Temperature , Joints/pathology , Liver/chemistry , Liver/metabolism , Mice , Mice, Inbred C3H
17.
Cancer Prev Res (Phila) ; 4(3): 329-46, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21372033

ABSTRACT

Elevated circulating estrogen levels are associated with increased risk of breast cancer in obese postmenopausal women. Following menopause, the biosynthesis of estrogens through CYP19 (aromatase)-mediated metabolism of androgen precursors occurs primarily in adipose tissue, and the resulting estrogens are then secreted into the systemic circulation. The potential links between obesity, inflammation, and aromatase expression are unknown. In both dietary and genetic models of obesity, we observed necrotic adipocytes surrounded by macrophages forming crown-like structures (CLS) in the mammary glands and visceral fat. The presence of CLS was associated with activation of NF-κB and increased levels of proinflammatory mediators (TNF-α, IL-1ß, Cox-2), which were paralleled by elevated levels of aromatase expression and activity in the mammary gland and visceral fat of obese mice. Analyses of the stromal-vascular and adipocyte fractions of the mammary gland suggested that macrophage-derived proinflammatory mediators induced aromatase and estrogen-dependent gene expression (PR, pS2) in adipocytes. Saturated fatty acids, which have been linked to obesity-related inflammation, stimulated NF-κB activity in macrophages leading to increased levels of TNF-α, IL-1ß, and Cox-2, each of which contributed to the induction of aromatase in preadipocytes. The discovery of the obesity → inflammation → aromatase axis in the mammary gland and visceral fat and its association with CLS may provide insight into mechanisms underlying the increased risk of hormone receptor-positive breast cancer in obese postmenopausal women, the reduced efficacy of aromatase inhibitors in the treatment of breast cancer in these women, and their generally worse outcomes. The presence of CLS may be a biomarker of increased breast cancer risk or poor prognosis.


Subject(s)
Aromatase/biosynthesis , Gene Expression Regulation, Neoplastic , Inflammation , Mammary Glands, Animal/metabolism , Obesity/metabolism , Adipocytes/cytology , Animals , Biomarkers, Tumor , Disease Models, Animal , Fatty Acids/metabolism , Female , Humans , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred C57BL
18.
J Immunol ; 186(5): 3076-84, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270404

ABSTRACT

The enzyme 5-lipoxygenase (5-LO) catalyzes the conversion of arachidonic acid into the leukotrienes, which are critical regulators of inflammation and inflammatory diseases, such as asthma and arthritis. Although leukotrienes are present in the synovial fluid of Lyme disease patients, their role in the development of Lyme arthritis has not been determined. In the current study, we used a murine model of Lyme arthritis to investigate the role 5-LO products might have in the development of this inflammatory disease. After infection of Lyme arthritis-susceptible C3H/HeJ mice with Borrelia burgdorferi, mRNA expression of 5-LO and 5-LO-activating protein was induced in the joints, and the 5-LO product leukotriene B(4) was produced. Using C3H 5-LO-deficient mice, we demonstrated that 5-LO activity was not necessary for the induction of Lyme arthritis, but that its deficiency resulted in earlier joint swelling and an inability to resolve arthritis as demonstrated by sustained arthritis pathology through day 60 postinfection. Although production of anti-Borrelia IgG was decreased in 5-LO-deficient mice, bacterial clearance from the joints was unaffected. Phagocytosis of B. burgdorferi and efferocytosis of apoptotic neutrophils was defective in macrophages from 5-LO-deficient mice, and uptake of opsonized spirochetes by neutrophils was reduced. These results demonstrate that products of the 5-LO metabolic pathway are not required for the development of disease in all models of arthritis and that caution should be used when targeting 5-LO as therapy for inflammatory diseases.


Subject(s)
Arachidonate 5-Lipoxygenase/deficiency , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Borrelia burgdorferi/immunology , Lyme Disease/enzymology , Lyme Disease/immunology , Animals , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/physiology , Arthritis, Experimental/enzymology , Cells, Cultured , Female , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/adverse effects , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Lyme Disease/pathology , Mice , Mice, 129 Strain , Mice, Inbred C3H , Mice, Knockout , Severity of Illness Index , Signal Transduction/genetics , Signal Transduction/immunology , Time Factors
19.
Infect Immun ; 78(11): 4593-600, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20823213

ABSTRACT

Deletion of the chemokine receptor CXCR2 prevents the recruitment of neutrophils into tissues and subsequent development of experimental Lyme arthritis. Following footpad inoculation of Borrelia burgdorferi, the agent of Lyme disease, expression of the CXCR2 ligand KC (CXCL1) is highly upregulated in the joints of arthritis-susceptible mice and is likely to play an important role in the recruitment of neutrophils to the site of infection. To test this hypothesis, we infected C3H KC(-/-) mice with B. burgdorferi and followed the development of arthritis and carditis. Ankle swelling was significantly attenuated during the peak of arthritis in the KC(-/-) mice. Arthritis severity scores were significantly lower in the KC(-/-) mice on days 11 and 21 postinfection, with fewer neutrophils present in the inflammatory lesions. Cardiac lesions were also significantly decreased in KC(-/-) mice at day 21 postinfection. There were, however, no differences between C3H wild-type and KC(-/-) mice in spirochete clearance from tissues. Two other CXCR2 ligands, LIX (CXCL5) and MIP-2 (CXCL2), were not increased to compensate for the loss of KC, and the production of several innate cytokines was unaltered. These results demonstrate that KC plays a critical nonredundant role in the development of experimental Lyme arthritis and carditis via CXCR2-mediated recruitment of neutrophils into the site of infection.


Subject(s)
Arthritis, Experimental/pathology , Borrelia burgdorferi/pathogenicity , Chemokine CXCL1/metabolism , Lyme Disease/pathology , Myocarditis/pathology , Neutrophil Infiltration/immunology , Receptors, Interleukin-8B/metabolism , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/microbiology , Borrelia burgdorferi/immunology , Disease Models, Animal , Female , Ligands , Lyme Disease/immunology , Lyme Disease/microbiology , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Myocarditis/immunology , Myocarditis/microbiology , Neutrophils/immunology , Severity of Illness Index , Signal Transduction
20.
J Exp Med ; 207(7): 1475-83, 2010 Jul 05.
Article in English | MEDLINE | ID: mdl-20584883

ABSTRACT

The sphingosine 1-phosphate receptor 1 (S1P(1)) promotes lymphocyte egress from lymphoid organs. Previous work showed that agonist-induced internalization of this G protein-coupled receptor correlates with inhibition of lymphocyte egress and results in lymphopenia. However, it is unclear if S1P(1) internalization is necessary for this effect. We characterize a knockin mouse (S1p1r(S5A/S5A)) in which the C-terminal serine-rich S1P(1) motif, which is important for S1P(1) internalization but dispensable for S1P(1) signaling, is mutated. T cells expressing the mutant S1P(1) showed delayed S1P(1) internalization and defective desensitization after agonist stimulation. Mutant mice exhibited significantly delayed lymphopenia after S1P(1) agonist administration or disruption of the vascular S1P gradient. Adoptive transfer experiments demonstrated that mutant S1P(1) expression in lymphocytes, rather than endothelial cells, facilitated this delay in lymphopenia. Thus, cell-surface residency of S1P(1) on T cells is a primary determinant of lymphocyte egress kinetics in vivo.


Subject(s)
Cell Membrane/metabolism , Chemotaxis, Leukocyte , Receptors, Lysosphingolipid/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Amino Acid Substitution/genetics , Animals , Base Sequence , Cell Membrane/drug effects , Chemotaxis, Leukocyte/drug effects , Endocytosis/drug effects , Fingolimod Hydrochloride , Kinetics , Lymphopenia/genetics , Lymphopenia/pathology , Lysophospholipids/agonists , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutant Proteins/metabolism , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/genetics , Sphingosine/agonists , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/pharmacology , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...