Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Blood ; 136(16): 1824-1836, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32483624

ABSTRACT

Yap1 and its paralogue Taz largely control epithelial tissue growth. We have identified that hematopoietic stem cell (HSC) fitness response to stress depends on Yap1 and Taz. Deletion of Yap1 and Taz induces a loss of HSC quiescence, symmetric self-renewal ability, and renders HSC more vulnerable to serial myeloablative 5-fluorouracil treatment. This effect depends on the predominant cytosolic polarization of Yap1 through a PDZ domain-mediated interaction with the scaffold Scribble. Scribble and Yap1 coordinate to control cytoplasmic Cdc42 activity and HSC fate determination in vivo. Deletion of Scribble disrupts Yap1 copolarization with Cdc42 and decreases Cdc42 activity, resulting in increased self-renewing HSC with competitive reconstitution advantages. These data suggest that Scribble/Yap1 copolarization is indispensable for Cdc42-dependent activity on HSC asymmetric division and fate. The combined loss of Scribble, Yap1, and Taz results in transcriptional upregulation of Rac-specific guanine nucleotide exchange factors, Rac activation, and HSC fitness restoration. Scribble links Cdc42 and the cytosolic functions of the Hippo signaling cascade in HSC fate determination.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , Membrane Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Biomarkers , Cell Proliferation , Cell Self Renewal , Cells, Cultured , Hematopoietic Stem Cells/cytology , Humans , Membrane Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
3.
Stem Cells Transl Med ; 8(6): 557-567, 2019 06.
Article in English | MEDLINE | ID: mdl-30793529

ABSTRACT

Bacterial and fungal infections are a major cause of morbidity and mortality in neutropenic patients. Donor-derived neutrophil transfusions have been used for prophylaxis or treatment for infection in neutropenic patients. However, the short half-life and the limited availability of large numbers of donor-derived neutrophils for transfusion remain a significant hurdle in the implementation of neutrophil transfusion therapy. Here, we investigate the in vitro and in vivo activity of neutrophils generated from human induced pluripotent stem cells (iPSC), a potentially unlimited resource to produce neutrophils for transfusion. Phenotypic analysis of iPSC-derived neutrophils reveal reactive oxygen species production at similar or slightly higher than normal peripheral blood neutrophils, but have an ∼50%-70% reduced Escherichia coli phagocytosis and phorbol 12-myristate 13-acetate induced formation of neutrophil extracellular traps (NET). Signaling of granulocytic precursors identified impaired AKT activation, but not ERK or STAT3, in agonist-stimulated iPSC-derived neutrophils. Expression of a constitutively activated AKT in iPSC-derived neutrophils restores most phagocytic activity and NET formation. In a model of bacterial induced peritonitis in immunodeficient mice, iPSC-derived neutrophils, with or without corrected AKT activation, migrate similarly to the peritoneal fluid as peripheral blood neutrophils, whereas the expression of activated AKT significantly improves their phagocytic activity in vivo. Stem Cells Translational Medicine 2019;8:557-567.


Subject(s)
Neutrophils/immunology , Phagocytosis , Adult , Animals , Escherichia coli/pathogenicity , Extracellular Traps/drug effects , Extracellular Traps/metabolism , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Neutrophils/cytology , Neutrophils/metabolism , Neutrophils/transplantation , Peritonitis/pathology , Peritonitis/therapy , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
4.
J Clin Invest ; 125(8): 3103-16, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26193632

ABSTRACT

Severe congenital neutropenia (SCN) is often associated with inherited heterozygous point mutations in ELANE, which encodes neutrophil elastase (NE). However, a lack of appropriate models to recapitulate SCN has substantially hampered the understanding of the genetic etiology and pathobiology of this disease. To this end, we generated both normal and SCN patient-derived induced pluripotent stem cells (iPSCs), and performed genome editing and differentiation protocols that recapitulate the major features of granulopoiesis. Pathogenesis of ELANE point mutations was the result of promyelocyte death and differentiation arrest, and was associated with NE mislocalization and activation of the unfolded protein response/ER stress (UPR/ER stress). Similarly, high-dose G-CSF (or downstream signaling through AKT/BCL2) rescues the dysgranulopoietic defect in SCN patient-derived iPSCs through C/EBPß-dependent emergency granulopoiesis. In contrast, sivelestat, an NE-specific small-molecule inhibitor, corrected dysgranulopoiesis by restoring normal intracellular NE localization in primary granules; ameliorating UPR/ER stress; increasing expression of CEBPA, but not CEBPB; and promoting promyelocyte survival and differentiation. Together, these data suggest that SCN disease pathogenesis includes NE mislocalization, which in turn triggers dysfunctional survival signaling and UPR/ER stress. This paradigm has the potential to be clinically exploited to achieve therapeutic responses using lower doses of G-CSF combined with targeting to correct NE mislocalization.


Subject(s)
Genetic Diseases, Inborn , Induced Pluripotent Stem Cells/enzymology , Leukocyte Elastase , Myelopoiesis/genetics , Neutropenia , Neutrophils/enzymology , Point Mutation , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Endoplasmic Reticulum Stress/genetics , Female , Genetic Diseases, Inborn/enzymology , Genetic Diseases, Inborn/genetics , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Precursor Cells/enzymology , Humans , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Male , Unfolded Protein Response/genetics
5.
Nat Commun ; 6: 5914, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25574809

ABSTRACT

Patients with organ failure of vascular origin have increased circulating haematopoietic stem cells and progenitors (HSC/P). Plasma levels of angiotensin II (Ang-II), are commonly increased in vasculopathies. Hyperangiotensinemia results in activation of a very distinct Ang-II receptor set, Rho family GTPase members, and actin in bone marrow endothelial cells (BMEC) and HSC/P, which results in decreased membrane integrin activation in both BMEC and HSC/P, and in HSC/P de-adhesion and mobilization. The Ang-II effect can be reversed pharmacologically and genetically by inhibiting Ang-II production or signalling through BMEC AT2R, HSCP Ang-II receptor type 1 (AT1R)/AT2R or HSC/P RhoA, but not by interfering with other vascular tone mediators. Hyperangiotensinemia and high counts of circulating HSC/P seen in sickle cell disease (SCD) as a result of vascular damage, is significantly decreased by Ang-II inhibitors. Our data define for the first time the role of Ang-II HSC/P traffic regulation and redefine the haematopoietic consequences of anti-angiotensin therapy in SCD.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Angiotensin II/metabolism , Cytoskeleton/metabolism , Hematopoietic Stem Cells/cytology , Stem Cells/cytology , Vascular Diseases/pathology , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Anemia, Sickle Cell/metabolism , Animals , Bone Marrow Cells/cytology , Cell Adhesion , Cell Membrane/metabolism , Crosses, Genetic , Endothelial Cells/cytology , Hematopoiesis , Humans , Integrin beta1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Nitric Oxide/chemistry , Signal Transduction
6.
Cell Rep ; 9(6): 2084-97, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25533346

ABSTRACT

In the bone marrow (BM), hematopoietic progenitors (HPs) reside in specific anatomical niches near osteoblasts (Obs), macrophages (MΦs), and other cells forming the BM microenvironment. A connection between immunosurveillance and traffic of HP has been demonstrated, but the regulatory signals that instruct the immune regulation of HP circulation are unknown. We discovered that the BM microenvironment deficiency of p62, an autophagy regulator and signal organizer, results in loss of autophagic repression of macrophage contact-dependent activation of Ob NF-κB signaling. Consequently, Ob p62-deficient mice lose bone, Ob Ccl4 expression, and HP chemotaxis toward Cxcl12, resulting in egress of short-term hematopoietic stem cells and myeloid progenitors. Finally, Ccl4 expression and myeloid progenitor egress are reversed by deficiency of the p62 PB1-binding partner Nbr1. A functional "MΦ-Ob niche" is required for myeloid progenitor/short-term stem cell retention, in which Ob p62 is required to maintain NF-κB signaling repression, osteogenesis, and BM progenitor retention.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Heat-Shock Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Macrophages/metabolism , Osteoblasts/metabolism , Osteogenesis , Signal Transduction , Stem Cell Niche , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy , Chemokine CCL4/metabolism , Heat-Shock Proteins/genetics , Hematopoietic Stem Cells/cytology , I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins , Macrophages/cytology , Mice , NF-kappa B/metabolism , Osteoblasts/cytology , Proteins/metabolism , Sequestosome-1 Protein
7.
Blood ; 123(4): 562-9, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24184683

ABSTRACT

Hereditary neutropenia is usually caused by heterozygous germline mutations in the ELANE gene encoding neutrophil elastase (NE). How mutations cause disease remains uncertain, but two hypotheses have been proposed. In one, ELANE mutations lead to mislocalization of NE. In the other, ELANE mutations disturb protein folding, inducing an unfolded protein response in the endoplasmic reticulum (ER). In this study, we describe new types of mutations that disrupt the translational start site. At first glance, they should block translation and are incompatible with either the mislocalization or misfolding hypotheses, which require mutant protein for pathogenicity. We find that start-site mutations, instead, force translation from downstream in-frame initiation codons, yielding amino-terminally truncated isoforms lacking ER-localizing (pre) and zymogen-maintaining (pro) sequences, yet retain essential catalytic residues. Patient-derived induced pluripotent stem cells recapitulate hematopoietic and molecular phenotypes. Expression of the amino-terminally deleted isoforms in vitro reduces myeloid cell clonogenic capacity. We define an internal ribosome entry site (IRES) within ELANE and demonstrate that adjacent mutations modulate IRES activity, independently of protein-coding sequence alterations. Some ELANE mutations, therefore, appear to cause neutropenia via the production of amino-terminally deleted NE isoforms rather than by altering the coding sequence of the full-length protein.


Subject(s)
Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Mutation , Neutropenia/metabolism , Protein Biosynthesis , Apoptosis , Codon , DNA Mutational Analysis , Endoplasmic Reticulum/metabolism , HL-60 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Neutrophils/cytology , Phenotype , Protein Denaturation , Protein Folding , Protein Isoforms/metabolism , U937 Cells
8.
Immunol Rev ; 256(1): 255-68, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24117826

ABSTRACT

The Rho family of guanosine triphosphatases (GTPases) is composed of members of the Ras superfamily of proteins. They are GTP-bound molecules with a modest intrinsic GTPase activity that can be accelerated upon activation/localization of specialized guanine nucleotide exchange factors. Members of this family act as molecular switches and are required for coordinated cytoskeletal rearrangements that are crucial in a set of specialized functions of mammalian stem cells. These functions include self-renewal, adhesion, and migration. Mouse gene-targeting studies have provided convincing evidence of the indispensable and dispensable roles of individual members of the Rho GTPase family and the putative upstream and downstream mediators in stem cell-specific functions. The role of Rho GTPases and related signaling pathways previously seen in other cell types and organisms have been confirmed in mammalian hematopoietic stem cells (HSCs), and new signaling pathways and unexpected functions unique to HSCs have been identified and dissected. This review summarizes our current understanding of the role of Rho family of GTPases on HSC and progenitor activity through cytoskeleton-mediated signaling pathways, providing insight about relevant signaling pathways that regulate mammalian stem cell self-renewal, adhesion, and migration.


Subject(s)
Cytoskeleton/physiology , Hematopoietic Stem Cells/physiology , rho GTP-Binding Proteins/metabolism , Animals , Hematopoiesis/physiology , Humans
9.
PLoS One ; 8(3): e59304, 2013.
Article in English | MEDLINE | ID: mdl-23555015

ABSTRACT

Recent studies have established that factor VIIa (FVIIa) binds to the endothelial cell protein C receptor (EPCR). FVIIa binding to EPCR may promote the endocytosis of this receptor/ligand complex. Rab GTPases are known to play a crucial role in the endocytic and exocytic pathways of receptors or receptor/ligand complexes. The present study was undertaken to investigate the role of Rab GTPases in the intracellular trafficking of EPCR and FVIIa. CHO-EPCR cells and human umbilical vein endothelial cells (HUVEC) were transduced with recombinant adenoviral vectors to express wild-type, constitutively active, or dominant negative mutant of various Rab GTPases. Cells were exposed to FVIIa conjugated with AF488 fluorescent probe (AF488-FVIIa), and intracellular trafficking of FVIIa, EPCR, and Rab proteins was evaluated by immunofluorescence confocal microscopy. In cells expressing wild-type or constitutively active Rab4A, internalized AF488-FVIIa accumulated in early/sorting endosomes and its entry into the recycling endosomal compartment (REC) was inhibited. Expression of constitutively active Rab5A induced large endosomal structures beneath the plasma membrane where EPCR and FVIIa accumulated. Dominant negative Rab5A inhibited the endocytosis of EPCR-FVIIa. Expression of constitutively active Rab11 resulted in retention of accumulated AF488-FVIIa in the REC, whereas expression of a dominant negative form of Rab11 led to accumulation of internalized FVIIa in the cytoplasm and prevented entry of internalized FVIIa into the REC. Expression of dominant negative Rab11 also inhibited the transport of FVIIa across the endothelium. Overall our data show that Rab GTPases regulate the internalization and intracellular trafficking of EPCR-FVIIa.


Subject(s)
Antigens, CD/genetics , Endocytosis/genetics , Factor VIIa/genetics , Receptors, Cell Surface/genetics , rab GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/genetics , Adenoviridae/genetics , Animals , Antigens, CD/metabolism , CHO Cells , Cricetinae , Endothelial Protein C Receptor , Factor VIIa/metabolism , Fluorescent Dyes , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Protein Transport/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , Transduction, Genetic , rab GTP-Binding Proteins/metabolism , rab4 GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism
10.
Front Immunol ; 4: 63, 2013.
Article in English | MEDLINE | ID: mdl-23493880

ABSTRACT

Multiple sclerosis (MS) is assumed to be an autoimmune disease initiated by autoreactive T cells that recognize central nervous system antigens. Although adaptive immunity is clearly involved in MS pathogenesis, innate immunity increasingly appears to be implicated in the disease. We and others have presented evidence that natural killer (NK) cells may be involved in immunoregulation in MS, leading to the question of whether a particular NK cell subtype will account for this effect. Changes of NK cell functionality in MS were associated with MS activity, and depletion of NK cells exacerbated the course of disease in a murine model of MS, experimental autoimmune encephalomyelitis. Several studies described a deficiency and transient "valleys" in NK cell killing activity in human MS, which may coincide with symptomatic relapse. However, the molecular basis of the defect in killing activity has not been determined. We discuss results on the expression of perforin in CD16(+) NK cells and the existence of an inverse relationship between myelin loaded phagocytes and the proportion of CD16(+) NK cells expressing perforin in the circulation. This inverse relationship is consistent with a role for NK cell killing activity in dampening autoimmunity. On the other hand, it has been broadly reported that first line MS therapies, such as interferon-beta, glatiramer acetate as well as escalation therapies such as fingolimod, daclizumab, or mitoxantrone seem to affect NK cell functionality and phenotype in vivo. Therefore, in this review we consider evidence for the immunoregulatory role of NK cells in MS and its animal models. Furthermore, we discuss the effect of MS treatments on NK cell activity.

11.
J Immunol ; 186(3): 1638-45, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21209279

ABSTRACT

Macrophages are a major component of the innate immune response, comprising the first line of defense against various intracellular pathogens, including Mycobacterium tuberculosis. In this report, we studied the factors that regulate growth of M. tuberculosis H37Rv in subpopulations of human monocyte-derived macrophages (MDMs). In healthy donors, M. tuberculosis H37Rv grew 5.6-fold more rapidly in CD14(hi) MDMs compared with that in CD14(lo)CD16(+) MDMs. Compared with CD14(lo)CD16(+) cells, M. tuberculosis H37Rv-stimulated CD14(hi) monocytes produced more IL-10 and had increased mRNA expression for c-Maf, a transcription factor that upregulates IL-10 gene expression. c-Maf small interfering RNA (siRNA) inhibited IL-10 production and growth of M. tuberculosis in CD14(hi) cells. Compared with CD14(lo)CD16(+) monocytes, M. tuberculosis H37Rv-stimulated CD14(hi) cells had increased expression of 22 genes whose promoters contained a c-Maf binding site, including hyaluronan synthase 1 (HAS1). c-Maf siRNA inhibited HAS1 expression in M. tuberculosis-stimulated CD14(hi) monocytes, and HAS1 siRNA inhibited growth of M. tuberculosis in CD14(hi) MDMs. M. tuberculosis H37Rv upregulated expression of HAS1 protein and its product, hyaluronan, in CD14(hi) MDMs. We conclude that M. tuberculosis grows more rapidly in CD14(hi) than in CD14(lo)CD16(+) MDMs because CD14(hi) cells have increased expression of c-Maf, which increases production of two key factors (hyaluronan and IL-10) that promote growth of M. tuberculosis.


Subject(s)
Lipopolysaccharide Receptors/biosynthesis , Macrophages/immunology , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Proto-Oncogene Proteins c-maf/physiology , Cell Differentiation/immunology , Cells, Cultured , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/biosynthesis , Humans , Hyaluronan Synthases , Hyaluronic Acid/physiology , Interleukin-10/biosynthesis , Interleukin-10/physiology , Macrophages/enzymology , Macrophages/metabolism , Monocytes/cytology , Monocytes/enzymology , Monocytes/immunology , Mycobacterium tuberculosis/pathogenicity , Proto-Oncogene Proteins c-maf/biosynthesis , Virulence/immunology
12.
Respir Res ; 11: 65, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20507598

ABSTRACT

BACKGROUND: Lung epithelial Na+ channels (ENaC) are regulated by cell Ca2+ signal, which may contribute to calcium antagonist-induced noncardiogenic lung edema. Although K+ channel modulators regulate ENaC activity in normal lungs, the therapeutical relevance and the underlying mechanisms have not been completely explored. We hypothesized that K+ channel openers may restore calcium channel blocker-inhibited alveolar fluid clearance (AFC) by up-regulating both apical and basolateral ion transport. METHODS: Verapamil-induced depression of heterologously expressed human alphabetagamma ENaC in Xenopus oocytes, apical and basolateral ion transport in monolayers of human lung epithelial cells (H441), and in vivo alveolar fluid clearance were measured, respectively, using the two-electrode voltage clamp, Ussing chamber, and BSA protein assays. Ca2+ signal in H441 cells was analyzed using Fluo 4AM. RESULTS: The rate of in vivo AFC was reduced significantly (40.6+/-6.3% of control, P<0.05, n=12) in mice intratracheally administrated verapamil. KCa3.1 (1-EBIO) and KATP (minoxidil) channel openers significantly recovered AFC. In addition to short-circuit current (Isc) in intact H441 monolayers, both apical and basolateral Isc levels were reduced by verapamil in permeabilized monolayers. Moreover, verapamil significantly altered Ca2+ signal evoked by ionomycin in H441 cells. Depletion of cytosolic Ca2+ in alphabetagamma ENaC-expressing oocytes completely abolished verapamil-induced inhibition. Intriguingly, KV (pyrithione-Na), K Ca3.1 (1-EBIO), and KATP (minoxidil) channel openers almost completely restored the verapamil-induced decrease in Isc levels by diversely up-regulating apical and basolateral Na+ and K+ transport pathways. CONCLUSIONS: Our observations demonstrate that K+ channel openers are capable of rescuing reduced vectorial Na+ transport across lung epithelial cells with impaired Ca2+ signal.


Subject(s)
Benzimidazoles/pharmacology , Calcium Channel Blockers/toxicity , Epithelial Cells/drug effects , Epithelial Sodium Channels/drug effects , Lung/drug effects , Minoxidil/pharmacology , Potassium Channels/agonists , Pulmonary Edema/drug therapy , Pyridines/pharmacology , Thiones/pharmacology , Verapamil/toxicity , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , KATP Channels/agonists , KATP Channels/metabolism , Lung/metabolism , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Potassium/metabolism , Potassium Channels/metabolism , Pulmonary Edema/chemically induced , Pulmonary Edema/metabolism , Sodium/metabolism , Xenopus laevis
13.
Blood ; 115(21): 4273-83, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20237315

ABSTRACT

Tissue factor (TF) on cell surfaces resides mostly in a cryptic state. It is not entirely clear how cryptic TF differs from procoagulantly active TF and how deencryption occurs. Here, we critically evaluated the importance of cystine 186-cystine 209 (Cys186-Cys209) bond formation for TF procoagulant activity and its de-encryption. Chinese hamster ovary cells transfected with TF(C186S), TF(C209S), or TF(C186S/C209S) expressed little procoagulant activity at the cell surface. TF monoclonal antibody and activated factor VII (FVIIa) binding studies showed that little TF protein was present at the cell surface in cells expressing mutant TF. Similar data were obtained in human umbilical vein endothelial cells (HUVECs) transduced to express TF(C186S), TF(C209S), or TF(C186S/C209S). Analysis of TF activity in HUVECs expressing similar levels of wild-type TF and TF(C186S/C209S) showed that TF mutant in the presence of saturating concentrations of FVIIa exhibited similar coagulant activity as that of wild-type TF. More importantly, treatment of HUVECs expressing TF(C186S/C209S) with HgCl(2) or ionomycin increased the cell-surface TF activity to the same extent as that of the wild-type TF. Our data provide clear evidence that TF lacking the Cys186-Cys209 bond is coagulantly active once it is complexed with FVIIa, and TF de-encryption does not require Cys186-Cys209 disulfide bond formation.


Subject(s)
Thromboplastin/chemistry , Animals , Base Sequence , CHO Cells , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Cricetinae , Cricetulus , Cystine/chemistry , DNA Primers/genetics , Dimerization , Disulfides/chemistry , Endothelial Cells/metabolism , Factor VIIa/metabolism , HL-60 Cells , Humans , Mercuric Chloride/pharmacology , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phosphatidylserines/metabolism , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thromboplastin/genetics , Thromboplastin/metabolism , Transfection
14.
J Immunol ; 183(10): 6639-45, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19864591

ABSTRACT

We determined whether human NK cells could contribute to immune defenses against Mycobacterium tuberculosis through production of IL-22. CD3(-)CD56(+) NK cells produced IL-22 when exposed to autologous monocytes and gamma-irradiated M. tuberculosis, and this depended on the presence of IL-15 and IL-23, but not IL-12 or IL-18. IL-15-stimulated NK cells expressed 10.6 times more DAP10 mRNA compared with control NK cells, and DAP10 siRNA inhibited IL-15-mediated IL-22 production by NK cells. Soluble factors produced by IL-15-activated NK cells inhibited growth of M. tuberculosis in macrophages, and this effect was reversed by anti-IL-22. Addition of rIL-22 to infected macrophages enhanced phagolysosomal fusion and reduced growth of M. tuberculosis. We conclude that NK cells can contribute to immune defenses against M. tuberculosis through production of IL-22, which inhibits intracellular mycobacterial growth by enhancing phagolysosomal fusion. IL-15 and DAP-10 elicit IL-22 production by NK cells in response to M. tuberculosis.


Subject(s)
Interleukins/immunology , Killer Cells, Natural/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Phagosomes/immunology , Tuberculosis/immunology , Humans , Interleukin-12/pharmacology , Interleukin-15/pharmacology , Interleukin-18/pharmacology , Interleukin-23/pharmacology , Interleukins/metabolism , Interleukins/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/microbiology , RNA, Messenger/agonists , RNA, Messenger/immunology , RNA, Messenger/metabolism , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism , Receptors, Immunologic/agonists , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Recombinant Proteins/pharmacology , Tuberculosis/microbiology , Interleukin-22
15.
Blood ; 114(9): 1974-86, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19587380

ABSTRACT

Although the binding of endothelial cell protein C receptor (EPCR) to its ligands is well characterized at the biochemical level, it remains unclear how EPCR interaction with its ligands at the cell surface impacts its cellular trafficking. We characterized the cellular localization and trafficking of EPCR in endothelial cells and a heterologous expression system. Immunofluorescence confocal microscopy studies revealed that a majority of EPCR is localized on the cell surface in membrane microdomains that are positive for caveolin-1. A small fraction of EPCR is also localized intracellularly in the recycling compartment. Factor VIIa (FVIIa) or activated protein C binding to EPCR promoted the internalization of EPCR. EPCR and EPCR-bound ligands were endocytosed rapidly via a dynamin- and caveolar-dependent pathway. The endocytosed receptor-ligand complexes were accumulated in a recycling compartment before being targeted back to the cell surface. EPCR-mediated FVIIa endocytosis/recycling also resulted in transport of FVIIa from the apical to the basal side. In vivo studies in mice showed that blockade of EPCR with EPCR-blocking antibodies impaired the early phase of FVIIa clearance. Overall, our results show that FVIIa or activated protein C binding to EPCR promotes EPCR endocytosis, and EPCR-mediated endocytosis may facilitate the transcytosis of FVIIa and its clearance from the circulation.


Subject(s)
Blood Coagulation Factors/biosynthesis , Factor VIIa/metabolism , Protein C/metabolism , Receptors, Cell Surface/biosynthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Endocytosis , Endothelium, Vascular/cytology , Humans , Ligands , Mice , Microscopy, Fluorescence/methods , Models, Biological , Protein Binding
16.
J Cell Sci ; 122(Pt 10): 1691-9, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19401334

ABSTRACT

In general, coronins play an important role in actin-based processes, and are expressed in a variety of eukaryotic cells, including Leishmania. Here, we show that Leishmania coronin preferentially distributes to the distal tip during cytokinesis, and interacts with microtubules through a microtubule-based motor, kinesin K39. We further show that reduction in coronin levels by 40-50% in heterozygous coronin mutants results in generation of bipolar cells (25-30%), specifically in the log phase, owing to unregulated growth of the corset microtubules. Further analysis of bipolar cells revealed that the main cause of generation of bipolar cell morphology is the intrusion of the persistently growing corset microtubules into the other daughter cell corset from the opposite direction. This defect in cytokinesis, however, disappears upon episomal gene complementation. Additionally, our attempts to prepare homozygous mutants were unsuccessful, as only the aneuploid cells survive the selection process. These results indicate that coronin regulates microtubule remodeling during Leishmania cytokinesis and is essentially required for survival of these parasites in culture.


Subject(s)
4-Butyrolactone/analogs & derivatives , Cytokinesis , Leishmania donovani/metabolism , Microtubules/metabolism , Protozoan Proteins/metabolism , 4-Butyrolactone/deficiency , 4-Butyrolactone/genetics , 4-Butyrolactone/metabolism , Aneuploidy , Animals , Antigens, Protozoan/metabolism , Cytokinesis/drug effects , Leishmania donovani/drug effects , Leishmania donovani/genetics , Leishmania donovani/growth & development , Leishmania donovani/ultrastructure , Macrolides/pharmacology , Microtubules/drug effects , Microtubules/ultrastructure , Mutation , Protozoan Proteins/genetics , Time Factors , Tubulin Modulators/pharmacology
17.
Retina ; 27(2): 211-5, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17290204

ABSTRACT

BACKGROUND: Antipericyte autoantibodies (APAAs) are present in high frequency among diabetic subjects with and without nonproliferative retinopathy. This study aimed to determine whether progression of retinopathy in type 2 diabetes was associated with the same medical risk factors in APAA-positive subjects as in APAA-negative subjects. METHODS: Type 2 diabetic patients with nonproliferative diabetic retinopathy at baseline were followed prospectively for 2 years monitoring progression of retinopathy. Thirty-eight (21.7%) of 175 patients had progression in Early Treatment Diabetic Retinopathy Study grade by > or =2 steps in at least 1 eye. Serum APAAs were detected by immunofluorescence on tissue-cultured bovine retinal pericytes. RESULTS: Progression of retinopathy was associated with HbA(1c) level (P = 0.002), diabetes duration (P = 0.03), and albumin/creatinine ratio (P = 0.02) in APAA-negative subjects but not in APAA-positive subjects. The association between progression and APAAs was strongest in the upper quartile for HbA(1c) level (>8.0%), where 71.4% of patients negative for APAAs had progression of retinopathy while only 24.1% of patients positive for APAAs had progression (P = 0.007). CONCLUSION: The results suggest that APAA presence is a modifier of risk of progression of retinopathy due to hyperglycemia and that it could be useful as a biochemical marker of risk of progression of diabetic retinopathy in type 2 diabetic patients with poor metabolic control.


Subject(s)
Autoantibodies/blood , Diabetes Mellitus, Type 2/immunology , Diabetic Retinopathy/immunology , Pericytes/immunology , Adult , Aged , Aged, 80 and over , Chromatography, High Pressure Liquid , Disease Progression , Female , Fluorescent Antibody Technique, Indirect , Glycated Hemoglobin/analysis , Humans , Male , Middle Aged , Prospective Studies , Retinal Neovascularization/immunology , Risk Factors
19.
Antioxid Redox Signal ; 7(11-12): 1486-93, 2005.
Article in English | MEDLINE | ID: mdl-16356112

ABSTRACT

Diabetic retinopathy is a sight-threatening complication of diabetes, and loss of pericytes represents early signs of its development. We tested the hypothesis that high glucose levels may induce signs of oxidative stress in cultured bovine retinal pericytes. Pericytes were exposed to either normal (5.5 mM) or high (22 mM) glucose levels for 1, 3, and 5 days. Signs of oxidative stress were measured by expression of copper/zinc superoxide dismutase, manganese superoxide dismutase, catalase, and glutathione peroxidase using real-time RTPCR. To elucidate the role of oxidative stress, we also measured glutathione (GSH) concentration in the cells and investigated the impact of thiol-reactive metal ions and hydrogen peroxide (H(2)O(2)) on intracellular GSH. Despite the stimulation with high glucose, thiol-reactive metal ions, or H(2)O(2), there was no clear increased expression of antioxidant enzymes or influence of GSH levels. Lipid peroxidation (malondialdehyde level) was increased in bovine aortic smooth muscle cells, but not in bovine retinal pericytes. The data indicate that pericytes do not develop oxidative stress in response to hyperglycemia. However, it is not definitively excluded that oxidative stress may occur after longer time periods of glucose stimulation.


Subject(s)
Glucose/pharmacology , Oxidative Stress/drug effects , Pericytes/drug effects , Pericytes/metabolism , Animals , Antioxidants/metabolism , Cattle , Cells, Cultured , Glutathione/metabolism , Malondialdehyde/metabolism , Muscle, Smooth/metabolism , RNA, Messenger/genetics , Superoxide Dismutase/metabolism , Transcription, Genetic/genetics
20.
Mol Biochem Parasitol ; 143(2): 152-64, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16024104

ABSTRACT

The presence of actin in Leishmania has recently been demonstrated, but the functional form of this protein (filamentous actin) has not yet been identified. We report here that the putative coronin homologue identified in the Leishmania genome is invariably associated with the filament-like structures of actin in Leishmania promastigotes. The occurrence of filamentous structures is significantly increased upon overexpression of Leishmania coronin as its GFP fusion product in Leishmania cells. However, expression of Leishmania actin or coronin alone in mammalian cells does not result in formation of any filament-like structures of Leishmania actin or association of Leishmania coronin with mammalian filamentous actin, but coexpression of both the proteins in these cells leads to formation of filamentous structures containing Leishmania actin and coronin. The high specificity of Leishmania coronin for Leishmania actin could be attributed to its unique structure as it differs from other coronins not only in the unique region but also in the actin-binding site and leucine zipper motif. These results taken together indicate that Leishmania contains a novel form of coronin which colocalizes with actin in filament-like structures in these cells.


Subject(s)
Actin Cytoskeleton/chemistry , Actins/analysis , Leishmania/chemistry , Microfilament Proteins/analysis , Actin Cytoskeleton/genetics , Actins/genetics , Amino Acid Sequence , Animals , Cell Line , Cricetinae , DNA, Protozoan/chemistry , DNA, Protozoan/genetics , Gene Expression , Leishmania/metabolism , Leishmania/ultrastructure , Leishmania donovani/chemistry , Leishmania donovani/metabolism , Leishmania donovani/ultrastructure , Leishmania major/chemistry , Leishmania major/metabolism , Leishmania major/ultrastructure , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Microscopy, Fluorescence , Microscopy, Immunoelectron , Molecular Sequence Data , Protein Binding , Protozoan Proteins/analysis , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...