Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 140(20): 2146-2153, 2022 11 17.
Article in English | MEDLINE | ID: mdl-35881848

ABSTRACT

Fetal/neonatal alloimmune thrombocytopenia (FNAIT) is a life-threatening bleeding disorder caused by maternal alloantibodies directed against paternally inherited human platelet alloantigens (HPAs) present on the surface of fetal and neonatal platelets. There are currently no approved therapies for the prevention of FNAIT. We report herein the ability of 2 human HPA-1a-specific therapeutic candidates, one a polyclonal, and the other a monoclonal antibody, to prevent alloimmunization in a novel preclinical mouse model of FNAIT. Both antibody preparations effected the rapid and complete elimination of HPA-1a+ platelets from circulation and prevented the development of HPA-1a alloantibodies. HPA-1a- female mice treated prophylactically with anti-HPA-1a antibody prior to exposure to HPA-1a+ platelets gave birth to HPA-1a+/- pups with significantly improved platelet counts and no bleeding symptoms. These preclinical data establish both the potential and threshold exposure targets for prophylactic treatment with HPA-1a-specific antibodies for the prevention of FNAIT in humans.


Subject(s)
Antigens, Human Platelet , Thrombocytopenia, Neonatal Alloimmune , Pregnancy , Humans , Female , Mice , Animals , Thrombocytopenia, Neonatal Alloimmune/prevention & control , Isoantibodies , Integrin beta3 , Prenatal Care , Fetus
2.
Pediatr Blood Cancer ; 71(4): e30835, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38212881

ABSTRACT

BACKGROUND: Fetal and neonatal alloimmune thrombocytopenia (FNAIT) ensues from parental incompatibility for platelet alloantigens with maternal sensitization. HPA-1a/1b incompatibility is the most common cause of FNAIT in Caucasians. Placental villitis and lower birthweight in FNAIT suggest anti-HPA-1a may have effects beyond inducing thrombocytopenia. OBJECTIVES: Does FNAIT secondary to anti-HPA-1a result in smaller newborns and, the corollary, does antenatal management of FNAIT increase birthweight? STUDY DESIGN: Birthweights of 270 FNAIT-affected newborns from a randomized clinical trial and a NAITbabies.org survey (135 paired siblings) were compared with those of published controls and treated to untreated FNAIT-affected siblings. Birthweights were converted to percentiles to account for gestational age, sex, and role of birth order in birth weight. Body weights of FNAIT-affected and -unaffected pups in a mouse FNAIT model were analyzed. RESULTS: Untreated siblings in both the clinical trial and NAITbabies.org cohorts were not small, compared with normal controls. However, treated siblings in both cohorts had significantly higher birthweight percentiles compared with their previous untreated affected sibling. After accounting for gestational age, sex, and birth order, increased birthweight percentile in treated compared with the untreated siblings remained significant in both cohorts. FNAIT-affected neonatal mice had lower bodyweights than FNAIT-unaffected pups. CONCLUSIONS: Untreated FNAIT-affected newborns were not small; however, treatment of FNAIT-affected pregnancies increased newborn birthweights despite corrections to account for other factors that might have influenced the results. High dose IVIG is believed to "block" FcRn and lower maternal anti-HPA-1a levels, and thus increase birthweights by reducing levels of maternal anti-HPA-1a and reducing placental villitis.


Subject(s)
Antigens, Human Platelet , Thrombocytopenia, Neonatal Alloimmune , Animals , Female , Humans , Infant, Newborn , Mice , Pregnancy , Birth Weight , Fetus , Gestational Age , Placenta , Thrombocytopenia, Neonatal Alloimmune/therapy , Male , Randomized Controlled Trials as Topic
3.
Arterioscler Thromb Vasc Biol ; 42(2): 193-204, 2022 02.
Article in English | MEDLINE | ID: mdl-34937389

ABSTRACT

OBJECTIVE: PECAM-1 (platelet endothelial cell adhesion molecule 1) is a 130 kDa member of the immunoglobulin (Ig) gene superfamily that is expressed on the surfaces of platelets and leukocytes and concentrated at the intercellular junctions of confluent endothelial cell monolayers. PECAM-1 Ig domains 1 and 2 (IgD1 and IgD2) engage in homophilic interactions that support a host of vascular functions, including support of leukocyte transendothelial migration and the maintenance of endothelial junctional integrity. The recently solved crystal structure of PECAM-1 IgD1 and IgD2 revealed a number of intermolecular interfaces predicted to play important roles in stabilizing PECAM-1/PECAM-1 homophilic interactions and in formation and maintenance of endothelial cell-cell contacts. We sought to determine whether the protein interfaces implicated in the crystal structure reflect physiologically important interactions. Approach and Results: We assessed the impact of single amino acid substitutions at the interfaces between opposing PECAM-1 molecules on homophilic binding and endothelial cell function. Substitution of key residues within the IgD1-IgD1 and IgD1-IgD2 interfaces but not those within the smaller IgD2-IgD2 interface, markedly disrupted PECAM-1 homophilic binding and its downstream effector functions, including the ability of PECAM-1 to localize at endothelial cell-cell borders, mediate the formation of endothelial tubes, and restore endothelial barrier integrity. CONCLUSIONS: Taken together, these results validate the recently described PECAM-1 IgD1/IgD2 crystal structure by demonstrating that specific residues visualized within the IgD1-IgD1 and IgD1-IgD2 interfaces of opposing molecules in the crystal are required for functionally important homophilic interactions. This information can now be exploited to modulate functions of PECAM-1 in vivo.


Subject(s)
Endothelial Cells/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Cell Adhesion , Cell Communication , Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells , Humans , Models, Molecular , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Protein Binding
4.
Genesis ; 58(2): e23346, 2020 02.
Article in English | MEDLINE | ID: mdl-31729819

ABSTRACT

Platelet endothelial cell adhesion molecule 1 (PECAM-1) is an adhesion and signaling receptor that is expressed on endothelial and hematopoietic cells and plays important roles in angiogenesis, vascular permeability, and regulation of cellular responsiveness. To better understanding the tissue specificity of PECAM-1 functions, we generated mice in which PECAM1, the gene encoding PECAM-1, could be conditionally knocked out. A targeting construct was created that contains loxP sites flanking PECAM1 exons 1 and 2 and a neomycin resistance gene flanked by flippase recognition target (FRT) sites that was positioned upstream of the 3' loxP site. The targeting construct was electroporated into C57BL/6 embryonic stem (ES) cells, and correctly targeted ES cells were injected into C57BL/6 blastocysts, which were implanted into pseudo-pregnant females. Resulting chimeric animals were bred with transgenic mice expressing Flippase 1 (FLP1) to remove the FRT-flanked neomycin resistance gene and mice heterozygous for the floxed PECAM1 allele were bred with each other to obtain homozygous PECAM1 flox/flox offspring, which expressed PECAM-1 at normal levels and had no overt phenotype. PECAM1 flox/flox mice were bred with mice expressing Cre recombinase under the control of the SRY-box containing gene 2 (Sox2Cre) promoter to delete the floxed PECAM1 allele in offspring (Sox2Cre;PECAM1 del/WT ), which were crossbred to generate Sox2Cre; PECAM1 del/del offspring. Sox2Cre; PECAM1 del/del mice recapitulated the phenotype of conventional global PECAM-1 knockout mice. PECAM1 flox/flox mice will be useful for studying distinct roles of PECAM-1 in tissue specific contexts and to gain insights into the roles that PECAM-1 plays in blood and vascular cell function.


Subject(s)
Gene Knockout Techniques/methods , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Animals , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
5.
Am J Physiol Cell Physiol ; 318(2): C392-C405, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31774702

ABSTRACT

Whether SLC4A11 transports ammonia and its potential mode of ammonia transport (NH4+, NH3, or NH3-2H+ transport have been proposed) are controversial. In the absence of ammonia, whether SLC4A11 mediates significant conductive H+(OH-) transport is also controversial. The present study was performed to determine the mechanism of human SLC4A11 ammonia transport and whether the transporter mediates conductive H+(OH-) transport in the absence of ammonia. We quantitated H+ flux by monitoring changes in intracellular pH (pHi) and measured whole cell currents in patch-clamp studies of HEK293 cells expressing the transporter in the absence and presence of NH4Cl. Our results demonstrate that SLC4A11 mediated conductive H+(OH-) transport that was stimulated by raising the extracellular pH (pHe). Ammonia-induced HEK293 whole cell currents were also stimulated by an increase in pHe. In studies using increasing NH4Cl concentrations with equal NH4+ extracellular and intracellular concentrations, the shift in the reversal potential (Erev) due to the addition of ammonia was compatible with NH3-H+ transport competing with H+(OH-) rather than NH3-nH+ (n ≥ 2) transport. The increase in equivalent H+(OH-) flux observed in the presence of a transcellular H+ gradient was also compatible with SLC4A11-mediated NH3-H+ flux. The NH3 versus Erev data fit a theoretical model suggesting that NH3-H+ and H+(OH-) competitively interact with the transporter. Studies of mutant SLC4A11 constructs in the putative SLC4A11 ion coordination site showed that both H+(OH-) transport and ammonia-induced whole cell currents were blocked suggesting that the H+(OH-) and NH3-H+ transport processes share common features involving the SLC4A11 transport mechanism.


Subject(s)
Ammonia/metabolism , Anion Transport Proteins/metabolism , Antiporters/metabolism , Ion Transport/physiology , Bicarbonates/metabolism , Cell Line , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Sodium/metabolism
6.
Eur J Immunol ; 47(1): 74-83, 2017 01.
Article in English | MEDLINE | ID: mdl-27759161

ABSTRACT

Pre-T cell receptor (TCR) signaling is required for pre-T cell survival, proliferation, and differentiation from the CD4 and CD8 double negative (DN) to the double positive (DP) stage. However, the pre-TCR signal transduction pathway is not fully understood and the signaling molecules involved have not been completely identified. Phospholipase Cγ (PLCγ) 1 is an important signaling molecule that generates two second messengers, diacylglycerol and inositol 1,4,5-trisphosphate, that are important to mediate PKC activation and intracellular Ca2+ flux in many signaling pathways. Previously, we have shown that PLCγ1 is important for TCR-mediated signaling, development and T-cell activation, but the role of PLCγ1 in pre-TCR signal transduction and pre-T cell development is not known. In this study, we demonstrated that PLCγ1 expression level in pre-T cells was comparable to that in mature T cells. Deletion of PLCγ1 prior to the pre-TCR signaling stage partially blocked the DN3 to DN4 transition and reduced thymic cellularity. We also demonstrated that deletion of PLCγ1 impaired pre-T cell proliferation without affecting cell survival. Further study showed that deficiency of PLCγ1 impaired pre-TCR mediated Ca2+ flux and Erk activation. Thus our studies demonstrate that PLCγ1 is important for pre-TCR mediated signal transduction and pre-T cell development.


Subject(s)
Cell Differentiation , Phospholipase C gamma/metabolism , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Animals , Biomarkers , Calcium/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cell Survival/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Genotype , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Phospholipase C gamma/deficiency , Phospholipase C gamma/genetics , Phosphorylation , Precursor Cells, T-Lymphoid/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism
7.
J Biol Chem ; 291(50): 26216-26225, 2016 Dec 09.
Article in English | MEDLINE | ID: mdl-27793989

ABSTRACT

Platelet Endothelial Cell Adhesion Molecule 1 (PECAM-1) is a major component of the endothelial cell intercellular junction. Previous studies have shown that PECAM-1 homophilic interactions, mediated by amino-terminal immunoglobulin homology domain 1, contribute to maintenance of the vascular permeability barrier and to its re-establishment following inflammatory or thrombotic insult. PECAM-1 glycans account for ∼30% of its molecular mass, and the newly solved crystal structure of human PECAM-1 immunoglobulin homology domain 1 reveals that a glycan emanating from the asparagine residue at position 25 (Asn-25) is located within the trans homophilic-binding interface, suggesting a role for an Asn-25-associated glycan in PECAM-1 homophilic interactions. In support of this possibility, unbiased molecular docking studies revealed that negatively charged α2,3 sialic acid moieties bind tightly to a groove within the PECAM-1 homophilic interface in an orientation that favors the formation of an electrostatic bridge with positively charged Lys-89, mutation of which has been shown previously to disrupt PECAM-1-mediated homophilic binding. To verify the contribution of the Asn-25 glycan to endothelial barrier function, we generated an N25Q mutant form of PECAM-1 that is not glycosylated at this position and examined its ability to contribute to vascular integrity in endothelial cell-like REN cells. Confocal microscopy showed that although N25Q PECAM-1 concentrates normally at cell-cell junctions, the ability of this mutant form of PECAM-1 to support re-establishment of a permeability barrier following disruption with thrombin was significantly compromised. Taken together, these data suggest that a sialic acid-containing glycan emanating from Asn-25 reinforces dynamic endothelial cell-cell interactions by stabilizing the PECAM-1 homophilic binding interface.


Subject(s)
Cell Communication/physiology , Endothelial Cells/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Polysaccharides/metabolism , Amino Acid Substitution , Cell Line , Endothelial Cells/cytology , Humans , Mutation, Missense , Platelet Endothelial Cell Adhesion Molecule-1/chemistry , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Polysaccharides/chemistry , Polysaccharides/genetics , Sialic Acids/chemistry , Sialic Acids/genetics , Sialic Acids/metabolism , Thrombin/chemistry , Thrombin/genetics , Thrombin/metabolism
8.
Am J Physiol Cell Physiol ; 311(5): C820-C830, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27581649

ABSTRACT

Congenital hereditary endothelial dystrophy (CHED), Harboyan syndrome (CHED with progressive sensorineural deafness), and potentially a subset of individuals with late-onset Fuchs' endothelial corneal dystrophy are caused by mutations in the SLC4A11 gene that results in corneal endothelial cell abnormalities. Originally classified as a borate transporter, the function of SLC4A11 as a transport protein remains poorly understood. Elucidating the transport function(s) of SLC4A11 is needed to better understand how its loss results in the aforementioned posterior corneal dystrophic disease processes. Quantitative PCR experiments demonstrated that, of the three known human NH2-terminal variants, SLC4A11-C is the major transcript expressed in human corneal endothelium. We studied the expression pattern of the three variants in mammalian HEK-293 cells and demonstrated that the SLC4A11-B and SLC4A11-C variants are plasma membrane proteins, whereas SLC4A11-A is localized intracellularly. SLC4A11-B and SLC4A11-C were shown to be multifunctional ion transporters capable of transporting H+ equivalents in both a Na+-independent and Na+-coupled mode. In both transport modes, SLC4A11-C H+ flux was significantly greater than SLC4A11-B. In the presence of ammonia, SLC4A11-B and SLC4A11-C generated inward currents that were comparable in magnitude. Chimera SLC4A11-C-NH2-terminus-SLC4A11-B experiments demonstrated that the SLC4A11-C NH2-terminus functions as an autoactivating domain, enhancing Na+-independent and Na+-coupled H+ flux without significantly affecting the electrogenic NH3-H(n)+ cotransport mode. All three modes of transport were significantly impaired in the presence of the CHED causing p.R109H (SLC4A11-C numbering) mutation. These complex ion transport properties need to be addressed in the context of corneal endothelial disease processes caused by mutations in SLC4A11.


Subject(s)
Anion Transport Proteins/genetics , Antiporters/genetics , Genetic Variation/genetics , Ion Transport/genetics , Cell Line , Cell Membrane/genetics , Corneal Dystrophies, Hereditary/genetics , Endothelium, Corneal/metabolism , HEK293 Cells , Hearing Loss, Sensorineural/genetics , Humans , Membrane Proteins/genetics , Middle Aged , Mutation/genetics
9.
J Biol Chem ; 290(9): 5391-404, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25568315

ABSTRACT

The extracellular loop 3 (EL-3) of SLC4 Na(+)-coupled transporters contains 4 highly conserved cysteines and multiple N-glycosylation consensus sites. In the electrogenic Na(+)-HCO3(-) cotransporter NBCe1-A, EL-3 is the largest extracellular loop and is predicted to consist of 82 amino acids. To determine the structural-functional importance of the conserved cysteines and the N-glycosylation sites in NBCe1-A EL-3, we analyzed the potential interplay between EL-3 disulfide bonding and N-glycosylation and their roles in EL-3 topological folding. Our results demonstrate that the 4 highly conserved cysteines form two intramolecular disulfide bonds, Cys(583)-Cys(585) and Cys(617)-Cys(642), respectively, that constrain EL-3 in a folded conformation. The formation of the second disulfide bond is spontaneous and unaffected by the N-glycosylation state of EL-3 or the first disulfide bond, whereas formation of the first disulfide bond relies on the presence of the second disulfide bond and is affected by N-glycosylation. Importantly, EL-3 from each monomer is adjacently located at the NBCe1-A dimeric interface. When the two disulfide bonds are missing, EL-3 adopts an extended conformation highly accessible to protease digestion. This unique adjacent parallel location of two symmetrically folded EL-3 loops from each monomer resembles a domain-like structure that is potentially important for NBCe1-A function in vivo. Moreover, the formation of this unique structure is critically dependent on the finely tuned interplay between disulfide bonding and N-glycosylation in the membrane processed NBCe1-A dimer.


Subject(s)
Cysteine/chemistry , Disulfides/chemistry , Protein Folding , Sodium-Bicarbonate Symporters/chemistry , Amino Acid Sequence , Binding Sites/genetics , Cysteine/genetics , Cysteine/metabolism , Disulfides/metabolism , Glycosylation , HEK293 Cells , Humans , Immunoblotting , Ion Transport/genetics , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Protein Multimerization , Protein Structure, Secondary , Sequence Homology, Amino Acid , Sodium-Bicarbonate Symporters/genetics , Sodium-Bicarbonate Symporters/metabolism
10.
Blood ; 123(6): 931-4, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24357731

ABSTRACT

Immune complexes consisting of heparin, platelet factor 4 (PF4), and PF4/heparin-reactive antibodies are central to the pathogenesis of heparin-induced thrombocytopenia (HIT). It is as yet unclear what triggers the initial induction of pathogenic antibodies. We identified B cells in peripheral blood of healthy adults that produce PF4/heparin-specific antibodies following in vitro stimulation with proinflammatory molecules containing deoxycytosine-deoxyguanosine (CpG). Similarly, B cells from unmanipulated wild-type mice produced PF4/heparin-specific antibodies following in vitro or in vivo CpG stimulation. Thus, both healthy humans and mice possess preexisting inactive/tolerant PF4/heparin-specific B cells. The findings suggest that breakdown of tolerance leads to PF4/heparin-specific B-cell activation and antibody production in patients developing HIT. Consistent with this concept, mice lacking protein kinase Cδ (PKCδ) that are prone to breakdown of B-cell tolerance produced anti-PF4/heparin antibodies spontaneously. Therefore, breakdown of tolerance can lead to PF4/heparin-specific antibody production, and B-cell tolerance may play an important role in HIT pathogenesis.


Subject(s)
Antibody Formation/immunology , Anticoagulants/adverse effects , B-Lymphocytes/immunology , Heparin/adverse effects , Platelet Factor 4/metabolism , Protein Kinase C-delta/physiology , Thrombocytopenia/immunology , Adult , Animals , Anticoagulants/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cells, Cultured , Heparin/metabolism , Humans , Immune Tolerance , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Factor 4/immunology , Prognosis , Thrombocytopenia/chemically induced , Thrombocytopenia/metabolism
11.
Pediatr Res ; 79(2): 318-24, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26492284

ABSTRACT

BACKGROUND: Postsurgical bleeding causes significant morbidity and mortality in children undergoing surgery for congenital heart defects (CHD). 22q11.2 deletion syndrome (DS) is the second most common genetic risk factor for CHD. The deleted segment of chromosome 22q11.2 encompasses the gene encoding glycoprotein (GP) Ibß, which is required for expression of the GPIb-V-IX complex on the platelet surface, where it functions as the receptor for von Willebrand factor (VWF). Binding of GPIb-V-IX to VWF is important for platelets to initiate hemostasis. It is not known whether hemizygosity for the gene encoding GPIbß increases the risk for bleeding following cardiac surgery for patients with 22q11.2 DS. METHODS: We performed a case-control study of 91 pediatric patients who underwent cardiac surgery with cardiopulmonary bypass from 2004 to 2012 at Children's Hospital of Wisconsin. RESULTS: Patients with 22q11.2 DS had larger platelets and lower platelet counts, bled more excessively, and received more transfusion support with packed red blood cells in the early postoperative period relative to control patients. CONCLUSION: Presurgical genetic testing for 22q11.2 DS may help to identify a subset of pediatric cardiac surgery patients who are at increased risk for excessive bleeding and who may require more transfusion support in the postoperative period.


Subject(s)
Cardiac Surgical Procedures/adverse effects , Chromosome Deletion , Chromosomes, Human, Pair 22 , DiGeorge Syndrome/genetics , Erythrocyte Transfusion/statistics & numerical data , Heart Defects, Congenital/surgery , Postoperative Hemorrhage/genetics , Postoperative Hemorrhage/therapy , Child , Child, Preschool , DiGeorge Syndrome/complications , DiGeorge Syndrome/diagnosis , Female , Genetic Association Studies , Genetic Predisposition to Disease , Heart Defects, Congenital/diagnosis , Hospitals, Pediatric , Humans , Infant , Infant, Newborn , Male , Phenotype , Postoperative Hemorrhage/diagnosis , Retrospective Studies , Risk Factors , Treatment Outcome , Wisconsin
12.
Am J Physiol Cell Physiol ; 308(2): C176-88, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25394471

ABSTRACT

The SLC4A11 gene mutations cause a variety of genetic corneal diseases, including congenital hereditary endothelial dystrophy 2 (CHED2), Harboyan syndrome, some cases of Fuchs' endothelial dystrophy (FECD), and possibly familial keratoconus. Three NH2-terminal variants of the human SLC4A11 gene, named SLC4A11-A, -B, and -C are known. The SLC4A11-B variant has been the focus of previous studies. Both the expression of the SLC4A11-C variant in the cornea and its functional properties have not been characterized, and therefore its potential pathophysiological role in corneal diseases remains to be explored. In the present study, we demonstrate that SLC4A11-C is the predominant SLC4A11 variant expressed in human corneal endothelial mRNA and that the transporter functions as an electrogenic H(+)(OH(-)) permeation pathway. Disulfonic stilbenes, including 4,4'-diisothiocyano-2,2'-stilbenedisulfonate (DIDS), 4,4'-diisothiocyanatodihydrostilbene-2,2'-disulfonate (H2DIDS), and 4-acetamido-4'-isothiocyanato-stilbene-2,2'-disulfonate (SITS), which are known to bind covalently, increased SLC4A11-C-mediated H(+)(OH(-)) flux by 150-200% without having a significant effect in mock-transfected cells. Noncovalently interacting 4,4'-diaminostilbene-2,2'-disulfonate (DADS) was without effect. We tested the efficacy of DIDS on the functionally impaired R109H mutant (SLC4A11-C numbering) that causes CHED2. DIDS (1 mM) increased H(+)(OH(-)) flux through the mutant transporter by ∼40-90%. These studies provide a basis for future testing of more specific chemically modified dilsulfonic stilbenes as potential therapeutic agents to improve the functional impairment of specific SLC4A11 mutant transporters.


Subject(s)
4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Anion Transport Proteins/metabolism , Antiporters/metabolism , Hydroxides/metabolism , Permeability/drug effects , Signal Transduction/drug effects , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/analogs & derivatives , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid/analogs & derivatives , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid/pharmacology , Anion Transport Proteins/genetics , Antiporters/genetics , Biological Transport/physiology , Cell Line , Cornea/drug effects , Cornea/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , HEK293 Cells , Humans , Mutation/genetics , RNA, Messenger/genetics
13.
J Autoimmun ; 56: 23-33, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25277651

ABSTRACT

CD31, a trans-homophilic inhibitory receptor expressed on both T- and B-lymphocytes, drives the mutual detachment of interacting leukocytes. Intriguingly, T cell CD31 molecules relocate to the immunological synapse (IS), where the T and B cells establish a stable interaction. Here, we show that intact CD31 molecules, which are able to drive an inhibitory signal, are concentrated at the periphery of the IS but are excluded from the center of the IS. At this site, were the cells establish the closest contact, the CD31 molecules are cleaved, and most of the extracellular portion of the protein, including the trans-homophilic binding sites, is shed from the cell surface. T cells lacking CD31 trans-homophilic binding sites easily establish stable interactions with B cells; at the opposite, CD31 signaling agonists inhibit T/B IS formation as well as the ensuing helper T cell activation and function. Confocal microscopy and flow cytometry analysis of experimental T/B IS shows that the T cell inhibitory effects of CD31 agonists depend on SHP-2 signaling, which reduces the phosphorylation of ZAP70. The analysis of synovial tissue biopsies from patients affected by rheumatoid arthritis showed that T cell CD31 molecules are excluded from the center of the T/B cell synapses in vivo. Interestingly, the administration of CD31 agonists in vivo significantly attenuated the development of the clinical signs of collagen-induced arthritis in DBA1/J mice. Altogether, our data indicate that the T cell co-inhibitory receptor CD31 prevents the formation of functional T/B immunological synapses and that therapeutic strategies aimed at sustaining CD31 signaling will attenuate the development of autoimmune responses in vivo.


Subject(s)
Arthritis, Experimental/immunology , Autoimmune Diseases/immunology , B-Lymphocytes/immunology , Immunological Synapses/immunology , Immunological Synapses/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Aged , Animals , Arthritis, Experimental/metabolism , Autoimmune Diseases/diagnosis , Autoimmune Diseases/metabolism , Biopsy , Cell Communication/drug effects , Cell Communication/immunology , Cell Line , Female , Humans , Lymphocyte Activation/immunology , Mice , Middle Aged , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction , Synovial Membrane/immunology , Synovial Membrane/pathology , T-Lymphocyte Subsets/drug effects , ZAP-70 Protein-Tyrosine Kinase/metabolism
14.
Blood ; 132(13): 1359-1360, 2018 09 27.
Article in English | MEDLINE | ID: mdl-30262581
15.
Blood ; 121(17): 3484-92, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23460609

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is an immune-mediated disorder that can cause fatal arterial or venous thrombosis/thromboembolism. Immune complexes consisting of platelet factor 4 (PF4), heparin, and PF4/heparin-reactive antibodies are central to the pathogenesis of HIT. However, the B-cell origin of HIT antibody production is not known. Here, we show that anti-PF4/heparin antibodies are readily generated in wild-type mice on challenge with PF4/heparin complexes, and that antibody production is severely impaired in B-cell-specific Notch2-deficient mice that lack marginal zone (MZ) B cells. As expected, Notch2-deficient mice responded normally to challenge with T-cell-dependent antigen nitrophenyl-chicken γ globulin but not to the T-cell-independent antigen trinitrophenyl-Ficoll. In addition, wild-type, but not Notch2-deficient, B cells plus B-cell-depleted wild-type splenocytes adoptively transferred into B-cell-deficient µMT mice responded to PF4/heparin complex challenge. PF4/heparin-specific antibodies produced by wild-type mice were IgG2b and IgG3 isotypes. An in vitro class-switching assay showed that MZ B cells were capable of producing antibodies of IgG2b and IgG3 isotypes. Lastly, MZ, but not follicular, B cells adoptively transferred into B-cell-deficient µMT mice responded to PF4/heparin complex challenge by producing PF4/heparin-specific antibodies of IgG2b and IgG3 isotypes. Taken together, these data demonstrate that MZ B cells are critical for PF4/heparin-specific antibody production.


Subject(s)
Antibody Formation , Autoantibodies/immunology , B-Lymphocytes/immunology , Heparin/immunology , Platelet Factor 4/immunology , Thrombocytopenia/immunology , Adoptive Transfer , Animals , Anticoagulants/adverse effects , Anticoagulants/immunology , Antigen-Presenting Cells/immunology , Autoantibodies/blood , B-Lymphocytes/chemistry , Coagulants/adverse effects , Coagulants/immunology , Flow Cytometry , Heparin/adverse effects , Immunization , Immunoglobulin Class Switching/immunology , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Factor 4/adverse effects , Receptor, Notch2/physiology , Thrombocytopenia/chemically induced , Thrombocytopenia/diagnosis
16.
Blood ; 121(10): 1858-67, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23264598

ABSTRACT

The integrin family is composed of a series of 24 αß heterodimer transmembrane adhesion receptors that mediate cell-cell and cell-extracellular matrix interactions. Adaptor molecules bearing immunoreceptor tyrosine-based activation motifs (ITAMs) have recently been shown to cooperate with specific integrins to increase the efficiency of transmitting ligand-binding-induced signals into cells. In human platelets, Fc receptor γ-chain IIa (FcγRIIa) has been identified as an ITAM-bearing transmembrane receptor responsible for mediating "outside-in" signaling through αIIbß3, the major adhesion receptor on the platelet surface. To explore the importance of FcγRIIa in thrombosis and hemostasis, we subjected FcγRIIa-negative and FcγRIIa-positive murine platelets to a number of well-accepted models of platelet function. Compared with their FcγRIIa-negative counterparts, FcγRIIa-positive platelets exhibited increased tyrosine phosphorylation of Syk and phospholipase Cγ2 and increased spreading upon interaction with immobilized fibrinogen, retracted a fibrin clot faster, and showed markedly enhanced thrombus formation when perfused over a collagen-coated flow chamber under conditions of arterial and venous shear. They also displayed increased thrombus formation and fibrin deposition in in vivo models of vascular injury. Taken together, these data establish FcγRIIa as a physiologically important functional conduit for αIIbß3-mediated outside-in signaling, and suggest that modulating the activity of this novel integrin/ITAM pair might be effective in controlling thrombosis.


Subject(s)
Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, IgG/physiology , Receptors, Immunologic/metabolism , Signal Transduction , Thrombosis/etiology , Tyrosine/metabolism , Animals , Arterioles/metabolism , Arterioles/pathology , Fibrin/metabolism , Fibrinogen/metabolism , Hemostasis/physiology , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Platelet Aggregation , Platelet Count , Thrombosis/metabolism , Thrombosis/pathology , Veins/metabolism , Veins/pathology
17.
J Biol Chem ; 288(11): 7894-7906, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23362273

ABSTRACT

In the kidney proximal tubule, NBCe1-A plays a critical role in absorbing HCO3(-) from cell to blood. NBCe1-A transmembrane segment 1 (TM1) is involved in forming part of the ion permeation pathway, and a missense mutation S427L in TM1 impairs ion transport, causing proximal renal tubular acidosis. In the present study, we examined the topology of NBCe1-A-TM1 in detail and its structural perturbation induced by S427L. We analyzed the N-terminal cytoplasmic region (Cys-389-Gln-424) of NBCe1-A-TM1 using the substituted cysteine scanning accessibility method combined with extensive chemical stripping, in situ chemical probing, and functional transport assays. NBCe1-A-TM1 was previously modeled on the anion exchanger 1 TM1 (AE1-TM1); however, our data demonstrated that the topology of AE1-TM1 differs significantly from NBCe1-A-TM1. Our findings revealed that NBCe1-A-TM1 is unusually long, consisting of 31 membrane-embedded amino acids (Phe-412 to Thr-442). The linker region (Arg-394-Pro-411) between the N terminus of TM1 and the cytoplasmic domain is minimally exposed to aqueous and is potentially folded in a helical structure that intimately interacts with the NBCe1-A cytoplasmic domain. In contrast, AE1-TM1 contains 25 amino acids connected to an aqueous-exposed cytoplasmic region. Based on our new NBCe1-A-TM1 model, Ser-427 resides in the middle of TM1. Leucine substitution at Ser-427 blocks the normal aqueous access to Thr-442, Ala-435, and Lys-404, implying a significant alteration of NBCe1-TM1 orientation. Our study provides novel structural insights into the pathogenic mechanism of S427L in mediating proximal renal tubular acidosis.


Subject(s)
Acidosis, Renal Tubular/genetics , Mutation , Sodium-Bicarbonate Symporters/chemistry , Acidosis, Renal Tubular/metabolism , Amino Acids/chemistry , Bicarbonates/chemistry , Biological Transport , Biotin/chemistry , Cell Membrane/metabolism , Cytoplasm/metabolism , HEK293 Cells , Humans , Lipid Bilayers/chemistry , Maleimides/chemistry , Mutagenesis, Site-Directed , Protein Binding , Protein Folding , Protein Structure, Tertiary , Sodium/metabolism , Sodium-Bicarbonate Symporters/metabolism
18.
Blood ; 119(8): 1935-45, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22210881

ABSTRACT

Platelets are essential for normal hemostasis, but close regulation is required to avoid the destructive effects of either inappropriate platelet activation or excessive responses to injury. Here, we describe a novel complex comprising the scaffold protein, spinophilin (SPL), and the tyrosine phosphatase, SHP-1, and show that it can modulate platelet activation by sequestering RGS10 and RGS18, 2 members of the regulator of G protein signaling family. We also show that SPL/RGS/SHP1 complexes are present in resting platelets where constitutive phosphorylation of SPL(Y398) creates an atypical binding site for SHP-1. Activation of the SHP-1 occurs on agonist-induced phosphorylation of SHP-1(Y536), triggering dephosphorylation and decay of the SPL/RGS/SHP1 complex. Preventing SHP-1 activation blocks decay of the complex and produces a gain of function. Conversely, deleting spinophilin in mice inhibits platelet activation. It also attenuates the rise in platelet cAMP normally caused by endothelial prostacyclin (PGI(2)). Thus, we propose that the role of the SPL/RGS/SHP1 complex in platelets is time and context dependent. Before injury, the complex helps maintain the quiescence of circulating platelets by maximizing the impact of PGI(2). After injury, the complex gradually releases RGS proteins, limiting platelet activation and providing a mechanism for temporal coordination of pro thrombotic and antithrombotic inputs.


Subject(s)
GTP-Binding Proteins/metabolism , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Platelet Activation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , RGS Proteins/metabolism , Animals , Binding Sites/genetics , Blood Platelets/metabolism , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Models, Biological , Mutation , Nerve Tissue Proteins/genetics , Phosphorylation , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , RGS Proteins/genetics , Signal Transduction , Transfection , Tyrosine/genetics , Tyrosine/metabolism
19.
Am J Physiol Cell Physiol ; 305(4): C392-405, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23636456

ABSTRACT

Mutations in SLC4A4, the gene encoding the electrogenic Na(+)-HCO3(-) cotransporter NBCe1, cause severe proximal renal tubular acidosis (pRTA), growth retardation, decreased IQ, and eye and teeth abnormalities. Among the known NBCe1 mutations, the disease-causing mechanism of the T485S (NBCe1-A numbering) mutation is intriguing because the substituted amino acid, serine, is structurally and chemically similar to threonine. In this study, we performed intracellular pH and whole cell patch-clamp measurements to investigate the base transport and electrogenic properties of NBCe1-A-T485S in mammalian HEK 293 cells. Our results demonstrated that Ser substitution of Thr485 decreased base transport by ~50%, and importantly, converted NBCe1-A from an electrogenic to an electroneutral transporter. Aqueous accessibility analysis using sulfhydryl reactive reagents indicated that Thr485 likely resides in an NBCe1-A ion interaction site. This critical location is also supported by the finding that G486R (a pRTA causing mutation) alters the position of Thr485 in NBCe1-A thereby impairing its transport function. By using NO3(-) as a surrogate ion for CO3(2-), our result indicated that NBCe1-A mediates electrogenic Na(+)-CO3(2-) cotransport when functioning with a 1:2 charge transport stoichiometry. In contrast, electroneutral NBCe1-T485S is unable to transport NO3(-), compatible with the hypothesis that it mediates Na(+)-HCO3(-) cotransport. In patients, NBCe1-A-T485S is predicted to transport Na(+)-HCO3(-) in the reverse direction from blood into proximal tubule cells thereby impairing transepithelial HCO3(-) absorption, possibly representing a new pathogenic mechanism for generating human pRTA.


Subject(s)
Acidosis, Renal Tubular/metabolism , Kidney Tubules, Proximal/metabolism , Mutation, Missense , Sodium-Bicarbonate Symporters/metabolism , Acidosis, Renal Tubular/genetics , Bicarbonates/metabolism , Carbonates/metabolism , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/pharmacology , Ethylmaleimide/pharmacology , Genetic Predisposition to Disease , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Kidney Tubules, Proximal/drug effects , Membrane Potentials , Mesylates/pharmacology , Mutagenesis, Site-Directed , Nitrates/metabolism , Patch-Clamp Techniques , Phenotype , Sodium/metabolism , Sodium-Bicarbonate Symporters/drug effects , Sodium-Bicarbonate Symporters/genetics , Transfection
20.
Biochemistry ; 52(15): 2597-608, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23418871

ABSTRACT

The activation state of many blood and vascular cells is tightly controlled by a delicate balance between receptors that contain immunoreceptor tyrosine-based activation motifs (ITAMs) and those that contain immunoreceptor tyrosine-based inhibitory motifs (ITIMs). Precisely how the timing of cellular activation by ITAM-coupled receptors is regulated by ITIM-containing receptors is, however, poorly understood. Using platelet endothelial cell adhesion molecule 1 (PECAM-1) as a prototypical ITIM-bearing receptor, we demonstrate that initiation of inhibitory signaling occurs via a novel, sequential process in which Src family kinases phosphorylate the C-terminal ITIM, thereby enabling phosphorylation of the N-terminal ITIM of PECAM-1 by other Src homology 2 domain-containing nonreceptor tyrosine kinases (NRTKs). NRTKs capable of mediating the second phosphorylation event include C-terminal Src kinase (Csk) and Bruton's tyrosine kinase (Btk). Btk and Csk function downstream of phosphatidylinositol 3-kinase (PI3K) activation during ITAM-dependent platelet activation. In ITAM-activated platelets that were treated with a PI3K inhibitor, PECAM-1 was phosphorylated but did not bind the tandem SH2 domain-containing tyrosine phosphatase SHP-2, indicating that it was not phosphorylated on its N-terminal ITIM. Csk bound to and phosphorylated PECAM-1 more efficiently than did Btk and required its SH2 domain to perform these functions. Additionally, the phosphorylation of the N-terminal ITIM of Siglec-9 by Csk is enhanced by the prior phosphorylation of its C-terminal ITIM, providing evidence that the ITIMs of other dual ITIM-containing receptors are also sequentially phosphorylated. On the basis of these findings, we propose that sequential ITIM phosphorylation provides a general mechanism for precise temporal control over the recruitment and activation of tandem SH2 domain-containing tyrosine phosphatases that dampen ITAM-dependent signals.


Subject(s)
Blood Platelets/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Signal Transduction , Agammaglobulinaemia Tyrosine Kinase , Amino Acid Motifs , CSK Tyrosine-Protein Kinase , Cytoplasm/metabolism , Humans , Phosphopeptides/metabolism , Phosphorylation , Platelet Membrane Glycoproteins/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , src Homology Domains , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL