Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
Front Immunol ; 12: 745854, 2021.
Article in English | MEDLINE | ID: mdl-34721415

ABSTRACT

Alpha 7 nicotinic acetylcholine receptor (α7 nAChR) is critical for the pathogenesis of Escherichia coli (E. coli) K1 meningitis, a severe central nervous system infection of the neonates. However, little is known about how E. coli K1 manipulates α7 nAChR signaling. Here, through employing immortalized cell lines, animal models, and human transcriptional analysis, we showed that E. coli K1 infection triggers releasing of secreted Ly6/Plaur domain containing 1 (SLURP1), an endogenous α7 nAChR ligand. Exogenous supplement of SLURP1, combined with SLURP1 knockdown or overexpression cell lines, showed that SLURP1 is required for E. coli K1 invasion and neutrophils migrating across the blood-brain barrier (BBB). Furthermore, we found that SLURP1 is required for E. coli K1-induced α7 nAChR activation. Finally, the promoting effects of SLURP1 on the pathogenesis of E. coli K1 meningitis was significantly abolished in the α7 nAChR knockout mice. These results reveal that E. coli K1 exploits SLURP1 to activate α7 nAChR and facilitate its pathogenesis, and blocking SLURP1-α7 nAChR interaction might represent a novel therapeutic strategy for E. coli K1 meningitis.


Subject(s)
Antigens, Ly/physiology , Blood-Brain Barrier , Escherichia coli Infections/microbiology , Escherichia coli/physiology , Meningitis, Escherichia coli/physiopathology , Urokinase-Type Plasminogen Activator/physiology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Antigens, Ly/genetics , Cell Line , Cerebrospinal Fluid/microbiology , Endothelial Cells/microbiology , Escherichia coli/isolation & purification , Hippocampus/metabolism , Host-Pathogen Interactions , Humans , Infant, Newborn , Memantine/pharmacology , Meningitis, Escherichia coli/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/physiology , Recombinant Proteins/metabolism , Specific Pathogen-Free Organisms , Urokinase-Type Plasminogen Activator/genetics , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , alpha7 Nicotinic Acetylcholine Receptor/deficiency
2.
Exp Eye Res ; 164: 118-128, 2017 11.
Article in English | MEDLINE | ID: mdl-28803936

ABSTRACT

Previously, we have reported that the Secreted Ly6/uPAR related protein-1 (SLURP1) serves an important immunomodulatory function in the ocular surface. Here, we examine the involvement of SLURP1 in regulating corneal angiogenic privilege. Slurp1 expression detected by QPCR, immunoblots and immunofluorescent stain, was significantly decreased in mouse corneas subjected to alkali burn-induced corneal neovascularization (CNV). Addition of exogenous SLURP1 (6XHis-tagged, E. coli expressed and partially purified using Ni-ion columns) significantly suppressed the tumor necrosis factor-α (TNF-α)-stimulated human umbilical cord vascular endothelial cell (HUVEC) tube formation. SLURP1 suppressed the HUVEC tube length, tube area and number of branch points, without affecting their viability and/or proliferation. Exogenous SLURP1 in HUVEC also suppressed the TNF-α-induced (i) interleukin-8 (IL-8) and TNF-α production, (ii) adhesion to different components of the extracellular matrix, (iii) migration, and (iv) nuclear localization of NFκB. Together, these results demonstrate that SLURP1 suppresses HUVEC tube formation by blocking nuclear translocation of NFκB, and suggest a potential role for SLURP1 in promoting corneal angiogenic privilege.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antigens, Ly/pharmacology , Corneal Neovascularization/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , NF-kappa B/metabolism , Urokinase-Type Plasminogen Activator/pharmacology , Animals , Antigens, Ly/physiology , Burns, Chemical/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Corneal Injuries/metabolism , Disease Models, Animal , Eye Burns/metabolism , Humans , Interleukin-8/metabolism , Mice , Tumor Necrosis Factor-alpha/metabolism , Urokinase-Type Plasminogen Activator/physiology
3.
J Exp Med ; 214(2): 475-489, 2017 02.
Article in English | MEDLINE | ID: mdl-28049627

ABSTRACT

Signaling lymphocytic activation molecule (SLAM)-associated protein (SAP) mutations in X-linked lymphoproliferative disease (XLP) lead to defective NKT cell development and impaired humoral immunity. Because of the redundancy of SLAM family receptors (SFRs) and the complexity of SAP actions, how SFRs and SAP mediate these processes remains elusive. Here, we examined NKT cell development and humoral immunity in mice completely deficient in SFR. We found that SFR deficiency severely impaired NKT cell development. In contrast to SAP deficiency, SFR deficiency caused no apparent defect in follicular helper T (TFH) cell differentiation. Intriguingly, the deletion of SFRs completely rescued the severe defect in TFH cell generation caused by SAP deficiency, whereas SFR deletion had a minimal effect on the defective NKT cell development in SAP-deficient mice. These findings suggest that SAP-dependent activating SFR signaling is essential for NKT cell selection; however, SFR signaling is inhibitory in SAP-deficient TFH cells. Thus, our current study revises our understanding of the mechanisms underlying T cell defects in patients with XLP.


Subject(s)
Natural Killer T-Cells/physiology , Signal Transduction/physiology , Signaling Lymphocytic Activation Molecule Family Member 1/physiology , Signaling Lymphocytic Activation Molecule Family/physiology , Animals , Antigens, Ly/physiology , CARD Signaling Adaptor Proteins/physiology , Immunity, Humoral , Kruppel-Like Transcription Factors/biosynthesis , Lymphoproliferative Disorders/genetics , Mice , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/physiology , Promyelocytic Leukemia Zinc Finger Protein , Signaling Lymphocytic Activation Molecule Associated Protein/physiology
4.
J Exp Med ; 214(2): 491-510, 2017 02.
Article in English | MEDLINE | ID: mdl-28057804

ABSTRACT

Natural killer (NK) cells are innate lymphoid cells with antitumor functions. Using an N-ethyl-N-nitrosourea (ENU)-induced mutagenesis screen in mice, we identified a strain with an NK cell deficiency caused by a hypomorphic mutation in the Bcl2 (B cell lymphoma 2) gene. Analysis of these mice and the conditional deletion of Bcl2 in NK cells revealed a nonredundant intrinsic requirement for BCL2 in NK cell survival. In these mice, NK cells in cycle were protected against apoptosis, and NK cell counts were restored in inflammatory conditions, suggesting a redundant role for BCL2 in proliferating NK cells. Consistent with this, cycling NK cells expressed higher MCL1 (myeloid cell leukemia 1) levels in both control and BCL2-null mice. Finally, we showed that deletion of BIM restored survival in BCL2-deficient but not MCL1-deficient NK cells. Overall, these data demonstrate an essential role for the binding of BCL2 to BIM in the survival of noncycling NK cells. They also favor a model in which MCL1 is the dominant survival protein in proliferating NK cells.


Subject(s)
Killer Cells, Natural/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Antigens, Ly/physiology , Bcl-2-Like Protein 11/physiology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Cycle , Cell Survival , Female , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Natural Cytotoxicity Triggering Receptor 1/physiology , Sulfonamides/pharmacology
5.
Biochem Biophys Res Commun ; 483(1): 590-595, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28011265

ABSTRACT

Roquin is an E3 ligase that regulates mRNA stability. Mice with a mutation in the Rc3h1 gene and Roquin protein, referred to as Roquinsan/san or sanroque mice, develop broad-spectrum chronic inflammatory conditions and autoimmune pathologies. Our laboratory recently reported that sanroque mice also develop extensive inflammation that is localized in the small intestine but is rare in the colon. Here, we demonstrate that small intestinal intraepithelial lymphocytes (IELs) are present in the epithelium of sanroque mice but that cell recoverability is low using standard extraction techniques even though lamina propria lymphocytes (LPLs) can be recovered in normal numbers. In studies aimed at characterizing T cell costimulatory markers and activation molecules on LPLs in sanroque mice, we identified Ly6C and 4-1BB (CD137) as being expressed at elevated levels on sanroque small intestinal LPLs, and we show that both of those subsets, in conjunction with cells expressing the KLRG1 T cell activation molecule, are sources of IL-17A, IFN-γ, and TNFα. TNFα was primarily produced by 4-1BB+, KLRG1-cells, but was also made by some 4-1BB-, KLRG1-cells, and 4-1BB-, KLRG1+ cells. These findings collectively suggest that the small intestinal inflammatory response in sanroque mice is driven, at least in part, by LPL activation through Ly6C and 4-1BB signaling, and they provide further evidence in support of using the sanroque mouse as an animal model of chronic small intestinal inflammation.


Subject(s)
Antigens, Ly/physiology , Lymphocytes/metabolism , Mucous Membrane/metabolism , Receptors, Immunologic/physiology , Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology , Animals , Crohn Disease/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Heterozygote , Inflammation , Interferon-gamma/metabolism , Interleukin-17/metabolism , Intestine, Small/metabolism , Lectins, C-Type , Mice , Tumor Necrosis Factor-alpha/metabolism
6.
J Hepatol ; 64(1): 128-34, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26341825

ABSTRACT

BACKGROUND & AIMS: NKp46(+) cells are major effector cells in the pathogenesis of hepatic ischemia reperfusion injury (IRI). Nevertheless, the precise role of unconventional subsets like the IL-22-producing NKp46(+) cells (NK22) remains unknown. The purpose of this study was to examine the role of NK22 cells in IRI in transplantation, particularly with respect to regulation by the transcription factor ROR-gamma-t (RORγt). METHODS: To explore the role of NK22 cells in IRI in the absence of adaptive immunity, B6.RORγt-(gfp/wt)-reporter and B6.RORγt-(gfp/gfp)-knockout (KO) mice on a Rag KO background underwent 90min partial warm ischemia, followed by 24h of reperfusion. RESULTS: Rag KO mice that possess fully functional NKp46(+) cells, and Rag-common-γ-chain-double-KO (Rag-γc-DKO) mice that lack T, B and NKp46(+) cells, were used as controls. We found that Rag-γc-DKO mice lacking NK22 cells show more severe levels of hepatocellular damage (GPT, histological injury) when compared to both Rag-RORγt-reporter and Rag KO mice that possess NK22 cells. Importantly, Rag-RORγt-reporter and Rag KO mice undergoing IRI expressed high protein levels of both IL-22 and GFP (RORγt), suggesting a protective role for RORγt(+) NK22 cells in IRI. Therefore, we tested the hypothesis that RORγt critically protects from IRI through the induction of hepatic NK22 cells by studying Rag-Rorγt-DKO mice under IRI conditions. We found that the lack of RORγt(+) NK22 cells in Rag-Rorγt-DKO mice significantly enhanced IR-induced hepatocellular injury, a phenotype that could be reversed upon adoptive transfer of Rag-Rorγt-reporter NK22 cells into DKO mice. CONCLUSIONS: RORγt(+) NK22 cells play an important protective role in IRI in mice.


Subject(s)
Antigens, Ly/physiology , Interleukins/biosynthesis , Liver/blood supply , Natural Cytotoxicity Triggering Receptor 1/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/physiology , Reperfusion Injury/prevention & control , Animals , Antigens, Ly/analysis , Homeodomain Proteins/physiology , Interferon-gamma/biosynthesis , Killer Cells, Natural/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/analysis , Nuclear Receptor Subfamily 1, Group F, Member 3/analysis , Reperfusion Injury/immunology , Interleukin-22
7.
J Exp Med ; 212(11): 1869-82, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26458769

ABSTRACT

Group 3 ILCs (ILC3s) are innate sources of IL-22 and IL-17 and include lymphoid tissue-inducer (LTi)-like and NKp46(+) subsets. Both depend on RORγt and aryl hydrocarbon receptor, but NKp46(+)ILC3s also require Notch and T-bet for their development and are transcriptionally distinct. The extent to which these subsets have unique functions, especially in the context of T cell- and B cell-sufficient mice, remains largely unclear. To investigate the specific function of NKp46(+)ILC3s among other ILC3 subsets and T cells, we generated mice selectively lacking NKp46(+)ILC3s or all ILC3s and crossed them to T cell-deficient mice, thus maintaining B cells in all mice. In mice lacking T cells, NKp46(+)ILC3s were sufficient to promote inflammatory monocyte accumulation in the anti-CD40 innate colitis model through marked production of GM-CSF. In T cell-competent mice, lack of NKp46(+)ILCs had no impact on control of intestinal C. rodentium infection, whereas lack of all ILC3s partially impaired bacterial control. Thus, NKp46(+)ILC3s have a unique capacity to promote inflammation through GM-CSF-induced accumulation of inflammatory monocytes, but are superseded by LTi-like ILC3s and T cells in controlling intestinal bacterial infection.


Subject(s)
Antigens, Ly/physiology , Colitis/immunology , Immunity, Innate , Lymphocytes/physiology , Natural Cytotoxicity Triggering Receptor 1/physiology , Adoptive Transfer , Animals , Antigens, Ly/analysis , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/physiology , Citrobacter rodentium , Enterobacteriaceae Infections/immunology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Interleukins/physiology , Male , Mice , Monocytes/physiology , Natural Cytotoxicity Triggering Receptor 1/analysis , Neutrophils/physiology , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/physiology , T-Lymphocytes/immunology , Interleukin-22
8.
Cardiovasc Res ; 108(1): 111-23, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26092098

ABSTRACT

AIMS: Macrophage apoptosis is a prominent feature of atherosclerosis, yet whether cell death-protected macrophages would favour the resolution of already established atherosclerotic lesions, and thus hold therapeutic potential, remains unknown. METHODS AND RESULTS: We irradiated then transplanted into Apoe(-/-) or LDLr(-/-) recipient mice harbouring established atherosclerotic lesions, bone marrow cells from mice displaying enhanced macrophage survival through overexpression of the antiapoptotic gene hBcl-2 (Mø-hBcl2 Apoe(-/-) or Mø-hBcl2 Apoe(+/+) LDLr(-/-)). Both recipient mice exhibited decreased lesional apoptotic cell content and reduced necrotic areas when repopulated with Mø-hBcl2 mouse-derived bone marrow cells. In contrast, only LDLr(-/-) recipients showed a reduction in plasma cholesterol levels and in atherosclerotic lesions. The absence of significant reduction of plasma cholesterol levels in the context of apoE deficiency highlighted macrophage-derived apoE as key in both the regulation of plasma and tissue cholesterol levels and the progression of pre-existing lesion. Accordingly, hBcl2 expression in macrophages was associated with larger pools of Kupffer cells and Ly-6C(low) monocytes, both high producers of apoE. Additionally, increased Kupffer cells population was associated with improved clearance of apoptotic cells and modified lipoproteins. CONCLUSION: Collectively, these data show that promoting macrophage survival provides a supplemental source of apoE, which hinders pre-existing plaque progression.


Subject(s)
Apolipoproteins E/physiology , Atherosclerosis/etiology , Macrophages/physiology , Animals , Antigens, Ly/physiology , Apoptosis , Cell Survival , Cholesterol/metabolism , Disease Progression , Male , Mice , Proto-Oncogene Proteins c-bcl-2/analysis , Receptors, LDL/physiology
9.
Exp Hematol ; 43(7): 578-85, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25892186

ABSTRACT

Identification and isolation of hematopoietic stem cells (HSCs) in mice is most commonly based on the expression of surface molecules Kit and Sca-1 and the absence of markers of mature lineages. However, Sca-1 is absent or weakly expressed in hematopoietic progenitors in many strains, including nonobese diabetic (NOD), BALB/c, C3H, and CBA mice. In addition, both Kit and Sca-1 levels are modulated following bone marrow injury. In these cases, other markers and dye exclusion methods have been employed to identify HSCs, yet there is no antibody-based stain that enables identification of HSCs and early progenitors when Kit and Sca-1 are inadequate. CD201 is a marker that is highly restricted to HSCs and progenitors, and CD27 is expressed at moderate-to-high levels on HSCs. We show here that combining CD201 and CD27 enables highly efficient isolation of long-term HSCs in NOD mice as well as in other strains, including SJL, FVB, AKR, BALB/c, C3H, and CBA. We also find that HSCs appear to maintain expression of CD201 and CD27 after hematopoietic injury when Kit expression is downregulated. These results suggest a widely applicable yet simple alternative for HSC isolation in settings where Kit and Sca-1 expression are insufficient.


Subject(s)
Blood Cells/chemistry , Cell Separation/methods , Flow Cytometry/methods , Hematopoietic Stem Cells/chemistry , Mice, Inbred Strains/blood , Receptors, Cell Surface/blood , Tumor Necrosis Factor Receptor Superfamily, Member 7/blood , Animals , Antigens, Ly/genetics , Antigens, Ly/physiology , Autoimmunity , Blood Cells/cytology , Bone Marrow/radiation effects , Cell Lineage , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Endothelial Protein C Receptor , Gene Expression , Hematopoietic Stem Cells/cytology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred NOD , Mice, Inbred Strains/genetics , Mice, Transgenic , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/physiology , Radiation Chimera , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/pathology , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
11.
Arterioscler Thromb Vasc Biol ; 34(6): 1209-20, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24723559

ABSTRACT

OBJECTIVE: The Cxcl12/Cxcr4 chemokine ligand/receptor axis mediates the mobilization of smooth muscle cell progenitors, driving injury-induced neointimal hyperplasia. This study aimed to investigate the role of endothelial Cxcr4 in neointima formation. APPROACH AND RESULTS: ß-Galactosidase staining using bone marrow x kinase (Bmx)-CreER(T2) reporter mice and double immunofluorescence revealed an efficient and endothelial-specific deletion of Cxcr4 in Bmx-CreER(T2+) compared with Bmx-CreER(T2-) Cxcr4-floxed apolipoprotein E-deficient (Apoe(-/-)) mice (referred to as Cxcr4(EC-KO)ApoE(-/-) and Cxcr4(EC-WT) ApoE(-/-), respectively). Endothelial Cxcr4 deficiency significantly increased wire injury-induced neointima formation in carotid arteries from Cxcr4(EC-KO)ApoE(-/-) mice. The lesions displayed a higher number of macrophages, whereas the smooth muscle cell and collagen content were reduced. This was associated with a significant reduction in reendothelialization and endothelial cell proliferation in injured Cxcr4(EC-KO)ApoE(-/-) carotids compared with Cxcr4(EC-WT)ApoE(-/-) controls. Furthermore, stimulation of human aortic endothelial cells with chemokine (C-X-C motif) ligand 12 (CXCL12) significantly enhanced their wound-healing capacity in an in vitro scratch assay, an effect that could be reversed with the CXCR4 antagonist AMD3100. Also, flow cytometric analysis showed a reduced mobilization of Sca1(+)Flk1(+)Cd31(+) and of Lin(-)Sca1(+) progenitors in Cxcr4(EC-KO) ApoE(-/-) mice after vascular injury, although Cxcr4 surface expression was unaltered. No differences could be detected in plasma concentrations of Cxcl12, vascular endothelial growth factor, sphingosine 1-phosphate, or Flt3 (fms-related tyrosine kinase 3) ligand, all cytokines with an established role in progenitor cell mobilization. Nonetheless, double immunofluorescence revealed a significant reduction in local endothelial Cxcl12 staining in injured carotids from Cxcr4(EC-KO)ApoE(-/-) mice. CONCLUSIONS: Endothelial Cxcr4 is crucial for efficient reendothelialization after vascular injury through endothelial wound healing and proliferation, and through the mobilization of Sca1(+)Flk1(+)Cd31(+) cells, often referred to as circulating endothelial progenitor cells.


Subject(s)
Atherosclerosis/pathology , Carotid Artery Injuries/pathology , Endothelial Cells/physiology , Neointima/pathology , Receptors, CXCR4/physiology , Animals , Antigens, Ly/physiology , Apolipoproteins E/physiology , Atherosclerosis/physiopathology , Cell Movement , Chemokine CXCL12/physiology , Female , Hyperplasia , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Protein-Tyrosine Kinases/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
12.
Circ Res ; 114(10): 1611-22, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24625784

ABSTRACT

RATIONALE: Healing after myocardial infarction involves the biphasic accumulation of inflammatory lymphocyte antigen 6C (Ly-6C)(high) and reparative Ly-6C(low) monocytes/macrophages (Mo/MΦ). According to 1 model, Mo/MΦ heterogeneity in the heart originates in the blood and involves the sequential recruitment of distinct monocyte subsets that differentiate to distinct macrophages. Alternatively, heterogeneity may arise in tissue from 1 circulating subset via local macrophage differentiation and polarization. The orphan nuclear hormone receptor, nuclear receptor subfamily 4, group a, member 1 (Nr4a1), is essential to Ly-6C(low) monocyte production but dispensable to Ly-6C(low) macrophage differentiation; dependence on Nr4a1 can thus discriminate between systemic and local origins of macrophage heterogeneity. OBJECTIVE: This study tested the role of Nr4a1 in myocardial infarction in the context of the 2 Mo/MΦ accumulation scenarios. METHODS AND RESULTS: We show that Ly-6C(high) monocytes infiltrate the infarcted myocardium and, unlike Ly-6C(low) monocytes, differentiate to cardiac macrophages. In the early, inflammatory phase of acute myocardial ischemic injury, Ly-6C(high) monocytes accrue in response to a brief C-C chemokine ligand 2 burst. In the second, reparative phase, accumulated Ly-6C(high) monocytes give rise to reparative Ly-6C(low) F4/80(high) macrophages that proliferate locally. In the absence of Nr4a1, Ly-6C(high) monocytes express heightened levels of C-C chemokine receptor 2 on their surface, avidly infiltrate the myocardium, and differentiate to abnormally inflammatory macrophages, which results in defective healing and compromised heart function. CONCLUSIONS: Ly-6C(high) monocytes orchestrate both inflammatory and reparative phases during myocardial infarction and depend on Nr4a1 to limit their influx and inflammatory cytokine expression.


Subject(s)
Antigens, Ly/physiology , Inflammation Mediators/physiology , Monocytes/metabolism , Myocardial Infarction/blood , Myocardial Infarction/prevention & control , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Animals , Antigens, Ly/blood , Cell Movement/physiology , Female , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Myocardial Infarction/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/blood
13.
Biol Reprod ; 90(3): 60, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24501175

ABSTRACT

A disintegrin and metallopeptidase domain 3 (ADAM3) is a sperm membrane protein reported to be critical for both sperm migration from the uterus into the oviduct in vivo and sperm binding to the zona pellucida in vitro. In order for ADAM3 to be expressed on the sperm surface, the interaction with testis-expressed gene 101 (TEX101), a glycosylphosphatidylinositol (GPI)-anchored protein, is essential. Without TEX101, ADAM3 is degraded during sperm transition through the epididymis. However, it is also known that TEX101 has to be shed and to disappear from testicular germ cells (TGCs) by the GPI-anchored protein-releasing activity of angiotensin-converting enzyme (ACE) for the correct localization of ADAM3 on the mature sperm surface to take place. Here, we found that in a mouse line with a disruption for another testis-specific GPI-anchored protein, lymphocyte antigen 6 complex, locus K (LY6K), the male mice became infertile and demonstrated a phenotype similar to that found in Adam3(-/-), Tex101(-/-), and Ace(-/-) mice. LY6K interacted with TEX101 and ADAM3 in the TGCs but disappeared from mature spermatozoa. Differing from more than 10 previously known gene knockout mouse lines that showed male infertility by impaired sperm migration into the oviduct, spermatozoa from Ly6K(-/-) mice had no aberrance in ADAM3. Thus, LY6K is a newly identified factor involved in sperm fertilizing ability. The lack of effect on ADAM3 in Ly6K(-/-) mice is indicative of an as yet undefined pathway in the mouse.


Subject(s)
Antigens, Ly/genetics , Antigens, Ly/physiology , Fallopian Tubes/physiology , Fertility/physiology , Glycosylphosphatidylinositols/chemistry , Sperm Motility/physiology , ADAM Proteins/genetics , ADAM Proteins/physiology , Animals , Blotting, Western , Calcium-Binding Proteins/genetics , Female , Fertilization in Vitro , Genetic Vectors , Hydrolysis , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Knockout , Molecular Chaperones/genetics , Pregnancy , Proteins/genetics , Proteins/physiology , Real-Time Polymerase Chain Reaction , Sperm-Ovum Interactions/drug effects , Sperm-Ovum Interactions/physiology , Transfection , Trypsin/chemistry
14.
J Leukoc Biol ; 95(6): 853-66, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24295829

ABSTRACT

STAT1 serves as an important regulator in the response to pathogens, oncogenic transformation, and genotoxic insults. It exerts these effects by shaping the innate and adaptive immune response and by participating in genotoxic stress pathways, leading to apoptosis and inhibition of cell proliferation. We have investigated the role of STAT1 in hematopoietic toxicity induced by doxorubicin in STAT1-proficient and -deficient mice. Whereas the early genotoxic effect of doxorubicin did not depend on STAT1, expression of STAT1 was required for efficient B lymphocyte repopulation in the recovery phase. We found a lower abundance of lymphocyte precursors in the BM of STAT1-deficient animals, which was particularly evident after doxorubicin-induced hematopoietic toxicity. In accordance, colony-forming assays with STAT1-deficient BM cells revealed a decreased number of pre-B colonies. Differentiation from the pro-B to the pre-B stage was not affected, as demonstrated by unaltered differentiation of purified B cell precursors from BM in the presence of IL-7. With the exception of Sca-1, expression of genes implicated in early lymphocyte development in pro-B cells did not depend on STAT1. Our findings indicate a specific requirement for STAT1 in lymphoid development before differentiation to pre-B cells, which becomes particularly apparent in the recovery phase from doxorubicin-induced hematopoietic toxicity.


Subject(s)
Antibiotics, Antineoplastic/toxicity , B-Lymphocytes/drug effects , Doxorubicin/toxicity , STAT1 Transcription Factor/physiology , Animals , Antigens, Ly/physiology , B-Lymphocytes/physiology , Cell Differentiation/drug effects , Cell Lineage , Hematopoiesis/drug effects , Interferon-gamma/physiology , Interleukin-7/pharmacology , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL
15.
PLoS One ; 8(8): e70610, 2013.
Article in English | MEDLINE | ID: mdl-23936457

ABSTRACT

We have shown that lithium treatment improves motor coordination in a spinocerebellar ataxia type 1 (SCA1) disease mouse model (Sca1(154Q/+)). To learn more about disease pathogenesis and molecular contributions to the neuroprotective effects of lithium, we investigated metabolomic profiles of cerebellar tissue and plasma from SCA1-model treated and untreated mice. Metabolomic analyses of wild-type and Sca1(154Q/+) mice, with and without lithium treatment, were performed using gas chromatography time-of-flight mass spectrometry and BinBase mass spectral annotations. We detected 416 metabolites, of which 130 were identified. We observed specific metabolic perturbations in Sca1(154Q/+) mice and major effects of lithium on metabolism, centrally and peripherally. Compared to wild-type, Sca1(154Q/+) cerebella metabolic profile revealed changes in glucose, lipids, and metabolites of the tricarboxylic acid cycle and purines. Fewer metabolic differences were noted in Sca1(154Q/+) mouse plasma versus wild-type. In both genotypes, the major lithium responses in cerebellum involved energy metabolism, purines, unsaturated free fatty acids, and aromatic and sulphur-containing amino acids. The largest metabolic difference with lithium was a 10-fold increase in ascorbate levels in wild-type cerebella (p<0.002), with lower threonate levels, a major ascorbate catabolite. In contrast, Sca1(154Q/+) mice that received lithium showed no elevated cerebellar ascorbate levels. Our data emphasize that lithium regulates a variety of metabolic pathways, including purine, oxidative stress and energy production pathways. The purine metabolite level, reduced in the Sca1(154Q/+) mice and restored upon lithium treatment, might relate to lithium neuroprotective properties.


Subject(s)
Antigens, Ly/physiology , Antipsychotic Agents/pharmacology , Biomarkers/metabolism , Cerebellum/metabolism , Disease Models, Animal , Lithium/pharmacology , Membrane Proteins/physiology , Metabolome/drug effects , Animals , Female , Gas Chromatography-Mass Spectrometry , Male , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Nat Med ; 19(6): 713-21, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23708291

ABSTRACT

The commensal flora can promote both immunity to pathogens and mucosal inflammation. How commensal-driven inflammation is regulated in the context of infection remains poorly understood. Here, we show that during acute mucosal infection of mice with Toxoplasma gondii, inflammatory monocytes acquire a tissue-specific regulatory phenotype associated with production of the lipid mediator prostaglandin E2 (PGE2). Notably, in response to commensals, inflammatory monocytes can directly inhibit neutrophil activation in a PGE2-dependent manner. Further, in the absence of inflammatory monocytes, mice develop severe neutrophil-mediated pathology in response to pathogen challenge that can be controlled by PGE2 analog treatment. Complementing these findings, inhibition of PGE2 led to enhanced neutrophil activation and host mortality after infection. These data demonstrate a previously unappreciated dual action of inflammatory monocytes in controlling pathogen expansion while limiting commensal-mediated damage to the gut. Collectively, our results place inflammatory monocyte-derived PGE2 at the center of a commensal-driven regulatory loop required to control host-commensal dialog during pathogen-induced inflammation.


Subject(s)
Gastrointestinal Diseases/immunology , Monocytes/immunology , Toxoplasmosis, Animal/immunology , Acute Disease , Animals , Antigens, Ly/physiology , Dinoprostone/biosynthesis , Female , Humans , Interleukin-10/biosynthesis , Mice , Mice, Inbred C57BL , Neutrophil Activation , Phenotype , Tumor Necrosis Factor-alpha/biosynthesis
17.
Exp Hematol ; 41(1): 113-23.e2, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22989761

ABSTRACT

Histone deacetylase inhibitors (HDIs) have been shown to enhance hematopoietic stem and progenitor cell activity and improve stem cell outcomes after ex vivo culture. Identification of gene targets of HDIs is required to understand the full potential of these compounds and can allow for improved stem cell culturing protocols. The molecular process that underlies the biological effects of valproic acid (VPA), a widely used HDI, on hematopoietic stem/progenitor cells was investigated by studying the early-response genes of VPA. These genes were linked to VPA-induced enhancement of cell function as measured by in vitro assays. Genome-wide gene expression studies revealed over-representation of genes involved in glutathione metabolism, receptor and signal transducer activity, and changes in the hematopoietic stem/progenitor cells surface profile after short, 24-hour VPA treatment. Sca-1, a well-known and widely used stem cell surface marker, was identified as a prominent VPA target. We showed that multiple HDIs induce Sca-1 expression on hematopoietic cells. VPA strongly preserved Sca-1 expression on Lin(-)Sca1(+)ckit(+) cells, but also reactivated Sca-1 on committed progenitor cells that were Sca-1(neg), thereby reverting them to the Lin(-)Sca1(+)ckit(+) phenotype. We demonstrated that reacquired Sca-1 expression coincided with induced self-renewal capacity as measured by in vitro replating assays, while Sca-1 itself was not required for the biological effects of VPA as demonstrated using Sca-1-deficient progenitor cells. In conclusion, our results show that VPA modulates several genes involved in multiple signal transduction pathways, of which Sca-1 was shown to mark cells with increased self-renewal capacity in response to HDIs.


Subject(s)
Antigens, Ly/physiology , Hematopoietic Stem Cells/drug effects , Histone Deacetylase Inhibitors/pharmacology , Membrane Proteins/physiology , Valproic Acid/pharmacology , Animals , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Female , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred C57BL
18.
Hypertension ; 60(3): 802-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22851736

ABSTRACT

Stem cell antigen (Sca) 1, a glycosyl phosphatidylinositol-anchored protein localized to lipid rafts, is upregulated in the heart during myocardial infarction and renovascular hypertension-induced cardiac hypertrophy. It has been suggested that Sca-1 plays an important role in myocardial infarction. To investigate the role of Sca-1 in cardiac hypertrophy, we performed aortic banding in Sca-1 cardiac-specific transgenic mice, Sca-1 knockout mice, and their wild-type littermates. Cardiac hypertrophy was evaluated by echocardiographic, hemodynamic, pathological, and molecular analyses. Sca-1 expression was upregulated and detected in cardiomyocytes after aortic banding surgery in wild-type mice. Sca-1 transgenic mice exhibited significantly attenuated cardiac hypertrophy and fibrosis and preserved cardiac function compared with wild-type mice after 4 weeks of aortic banding. Conversely, Sca-1 knockout dramatically worsened cardiac hypertrophy, fibrosis, and dysfunction after pressure overload. Furthermore, aortic banding-induced activation of Src, mitogen-activated protein kinases, and Akt was blunted by Sca-1 overexpression and enhanced by Sca-1 deficiency. Our results suggest that Sca-1 protects against cardiac hypertrophy and fibrosis via regulation of multiple pathways in cardiomyocytes.


Subject(s)
Antigens, Ly/physiology , Cardiomegaly/prevention & control , Cardiomegaly/physiopathology , Heart/physiopathology , Hypertension/physiopathology , Membrane Proteins/physiology , Myocardium/pathology , Animals , Antigens, Ly/genetics , Cardiomegaly/diagnostic imaging , Echocardiography , Fibrosis/physiopathology , Fibrosis/prevention & control , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Animal , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Up-Regulation , src-Family Kinases/metabolism
19.
Anesthesiology ; 117(3): 602-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22820848

ABSTRACT

BACKGROUND: Neutrophils are one of the predominant immune cells initially migrating to surgical wound edges. They produce mediators both associated with supporting (interleukin [IL]-1ß, C5a) and reducing (opioid peptides) pain. Studies demonstrate neutrophil depletion/blockade reduces nociceptive sensitization after nerve injury and carrageenan administration, but enhance sensitization in complete Freund's adjuvant inflammation. This research identifies the contribution of infiltrating neutrophils to incisional pain and inflammation. METHODS: Antibody-mediated Gr1 neutrophil depletion preceded hind paw incisions. Sensitization to mechanical and thermal stimuli, effects on edema and local levels of IL-1ß and C5a were measured. Local effects of C5a or IL-1 receptor antagonists PMX-53 and anakinra on sensitization after neutrophil depletion were examined. Groups of 4-8 mice were used. RESULTS: Anti-Gr1 antibody depleted more than 90% of circulating and infiltrating skin neutrophils after incision. Neutrophil depletion did not change magnitude or duration of mechanical hypersensitivity in incised mice. However, paw edema was significantly reduced and heat hypersensitivity was slightly increased in depleted animals. In depleted animals IL-1ß levels were half of controls 24 h after incision, whereas C5a levels were increased in both. Prominent IL-1ß immunohistochemical staining of epidermis was seen in both groups. PMX-53 and anakinra reduced incisional mechanical and heat nociceptive sensitization to the same extent, regardless of neutrophil depletion. CONCLUSIONS: Neutrophil-derived IL-1ß and C5a do not appear to contribute critically to peri-incisional nociceptive signaling. Other sources of mediators, such as epidermal cells, may need to be considered. Controlling inflammatory activation of resident cells in epidermis/deeper structures may show therapeutic efficacy in reducing pain from surgical incisions.


Subject(s)
Inflammation/etiology , Neutrophils/physiology , Pain/physiopathology , Receptors, Chemokine/physiology , Animals , Antigens, Ly/physiology , Complement C5a/analysis , Interleukin-1beta/analysis , Male , Mice , Mice, Inbred C57BL , Receptors, Chemokine/analysis
20.
Immunity ; 36(6): 986-1002, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22683125

ABSTRACT

Humans and mice deficient in the adaptor protein SAP (Sh2d1a) have a major defect in humoral immunity, resulting from a lack of T cell help for B cells. The role of SAP in this process is incompletely understood. We found that deletion of receptor Ly108 (Slamf6) in CD4(+) T cells reversed the Sh2d1a(-/-) phenotype, eliminating the SAP requirement for germinal centers. This potent negative signaling by Ly108 required immunotyrosine switch motifs (ITSMs) and SHP-1 recruitment, resulting in high amounts of SHP-1 at the T cell:B cell synapse, limiting T cell:B cell adhesion. Ly108-negative signaling was important not only in CD4(+) T cells; we found that NKT cell differentiation was substantially restored in Slamf6(-/-)Sh2d1a(-/-) mice. The ability of SAP to regulate both positive and negative signals in T cells can explain the severity of SAP deficiency and highlights the importance of SAP and SHP-1 competition for Ly108 ITSM binding as a rheostat for the magnitude of T cell help to B cells.


Subject(s)
Antigens, Ly/physiology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Cooperation/physiology , Lymphopoiesis/physiology , Natural Killer T-Cells/cytology , Amino Acid Motifs , Animals , Antigens, Ly/genetics , Germinal Center/immunology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunological Synapses/immunology , Inositol Polyphosphate 5-Phosphatases , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphoric Monoester Hydrolases/physiology , Phosphorylation , Phosphotyrosine/physiology , Protein Processing, Post-Translational , Signaling Lymphocytic Activation Molecule Associated Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...