Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
J Thromb Haemost ; 17(1): 206-219, 2019 01.
Article in English | MEDLINE | ID: mdl-30388316

ABSTRACT

Essentials Acquired Glanzmann thrombasthenia (aGT) is generally caused by function-blocking antibodies (Abs). We demonstrated a unique aGT case due to marked reduction of αIIbß3 with anti-αIIbß3 Abs. The anti-αIIbß3 Abs of the patient did not inhibit platelet function but reduced surface αIIbß3. Internalization of αIIbß3 induced by the Abs binding may be responsible for the phenotype. SUMMARY: Background Acquired Glanzmann thrombasthenia (aGT) is a bleeding disorder generally caused by function-blocking anti-αIIbß3 autoantibodies. Aim We characterize an unusual case of aGT caused by marked reduction of surface αIIbß3 with non-function-blocking anti-αIIbß3 antibodies (Abs). Methods A 72-year-old male suffering from immune thrombocytopenia since his 50s showed exacerbation of bleeding symptom despite mild thrombocytopenia. Platelet aggregation was absent with all agonists but ristocetin. Analysis of αIIbß3 expression and genetic analysis were performed. We also analyzed effects of anti-αIIbß3 Abs of the patient on platelet function and αIIbß3 expression. Results Surface αIIbß3 expression was markedly reduced to around 5% of normal, whereas his platelets contained αIIbß3 to the amount of 40-50% of normal. A substantial amount of fibrinogen was also detected in his platelets. There were no abnormalities in ITGA2B and ITGB3 cDNA. These results indicated that reduced surface αIIbß3 expression caused a GT phenotype, and active internalization of αIIbß3 was suggested. Anti-αIIbß3 IgG Abs were detected in platelet eluate and plasma. These Abs did not inhibit PAC-1 binding, indicating that the Abs were non-function-blocking. Surface αIIbß3 expression of a megakaryocytic cell line and cultured megakaryocytes tended to be impaired by incubation with the patient's Abs. After 2 years of aGT diagnosis, his bleeding symptom improved and surface αIIbß3 expression was recovered to 20% of normal with reduction of anti-αIIbß3 Abs. Conclusion We demonstrated a unique aGT phenotype due to marked reduction of surface αIIbß3. Internalization induced by anti-αIIbß3 Abs may be responsible in part for the phenotype.


Subject(s)
Autoantibodies/immunology , Blood Platelets/immunology , Integrin alpha2/immunology , Integrin beta3/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Thrombasthenia/immunology , Aged , Blood Platelets/metabolism , Cells, Cultured , Epistaxis/blood , Epistaxis/immunology , Gastrointestinal Hemorrhage/blood , Gastrointestinal Hemorrhage/immunology , Humans , Integrin alpha2/blood , Integrin beta3/blood , Male , Phenotype , Platelet Function Tests , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombasthenia/blood , Thrombasthenia/diagnosis
2.
Vox Sang ; 108(3): 243-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25536173

ABSTRACT

BACKGROUND AND OBJECTIVES: This study compares the frequency of adverse transfusion reactions (ATRs) after first transfusions with the frequency of ATRs for subsequent (non-first) transfusions. MATERIALS AND METHODS: Five hospitals agreed to systematically collect and share 2 years of data. This was a retrospective observational analysis of data including the number of transfusion episodes and ATRs for red blood cells (RBCs), fresh frozen plasma (FFP) and platelet concentrates (PCs) given to first-time transfusion recipients and to those previously transfused. RESULTS: First transfusion ATRs to RBCs, FFP and PCs were 1.08%, 2.84% and 3.34%, respectively. These are higher than ATR incidences to RBCs (0.69%), FFP (1.91%) and PCs (2.75%) on subsequent transfusions. Specifically, first transfusion incidences of febrile non-haemolytic transfusion reactions (FNHTRs) to RBCs (0.43%) and allergic reactions to FFP (2.51%) were higher than on subsequent transfusions (RBCs: 0.23%, FFP: 1.65%). CONCLUSION: There are risks of ATRs on the first transfusion as well as transfusions of patients with transfusion history.


Subject(s)
Blood Transfusion/statistics & numerical data , Transfusion Reaction/epidemiology , Female , Humans , Incidence , Male , Retrospective Studies , Transfusion Reaction/etiology
3.
Cell Death Differ ; 19(11): 1856-69, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22790873

ABSTRACT

Anti-apoptotic Bcl-2 family proteins, which inhibit the mitochondrial pathway of apoptosis, are involved in the survival of various hematopoietic lineages and are often dysregulated in hematopoietic malignancies. However, their involvement in the megakaryocytic lineage is not well understood. In the present paper, we describe the crucial anti-apoptotic role of Mcl-1 and Bcl-xL in this lineage at multistages. The megakaryocytic lineage-specific deletion of both, in sharp contrast to only one of them, caused apoptotic loss of mature megakaryocytes in the fetal liver and systemic hemorrhage, leading to embryonic lethality. ABT-737, a Bcl-xL/Bcl-2/Bcl-w inhibitor, only caused thrombocytopenia in adult wild-type mice, but further induced massive mature megakaryocyte apoptosis in the Mcl-1 knockout mice, leading to severe hemorrhagic anemia. All these phenotypes were fully restored if Bak and Bax, downstream apoptosis executioners, were also deficient. In-vitro study revealed that the Jak pathway maintained Mcl-1 and Bcl-xL expression levels, preventing megakaryoblastic cell apoptosis. Similarly, both were involved in reticulated platelet survival, whereas platelet survival was dependent on Bcl-xL due to rapid proteasomal degradation of Mcl-1. In conclusion, Mcl-1 and Bcl-xL regulate the survival of the megakaryocytic lineage, which is critically important for preventing lethal or severe hemorrhage in both developing and adult mice.


Subject(s)
Apoptosis , Megakaryocytes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Animals , Biphenyl Compounds/pharmacology , Cell Line , Cell Lineage , Humans , Janus Kinases/metabolism , Megakaryocytes/drug effects , Mice , Mice, Knockout , Myeloid Cell Leukemia Sequence 1 Protein , Nitrophenols/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors
4.
J Thromb Haemost ; 10(5): 799-806, 2012 May.
Article in English | MEDLINE | ID: mdl-22409309

ABSTRACT

BACKGROUND: Eltrombopag is an oral, non-peptide thrombopoietin receptor agonist that has shown efficacy and safety in chronic immune thrombocytopenia (ITP). However, ethnic differences in eltrombopag exposure have been reported: area under the curve exposure to eltrombopag was 87% greater among ITP patients of East Asian descent than among ITP patients of non-East Asian ITP descent. OBJECTIVES: To evaluate the efficacy and safety of eltrombopag by using, in Japanese ITP patients, lower starting (12.5 mg) and maximum (50 mg) doses of eltrombopag than the standard starting (50 mg) and maximum (75 mg) doses approved in the USA and Europe. PATIENTS: We examined 23 Japanese patients with previously treated chronic ITP with a platelet count of < 30,000 µL(-1) in a multicenter study comprising a randomized, double-blind, placebo-controlled phase for 6-week evaluation (15 eltrombopag, and eight placebo) and an open-label phase for 6-month evaluation (23 eltrombopag). RESULTS AND CONCLUSIONS: The response rate (platelet count of ≥ 50,000 µL(-1) ) at week 6 of the 6-week double-blind phase was 60% in eltrombopag-treated patients and 0% in placebo-treated patients. Ten of 23 patients (43.5%) responded for ≥ 75% of predefined assessment visits during the 6-month open-label phase. Notably, 22% (5/23) of patients responded to 12.5 mg of eltrombopag, which was administered within the first 3 weeks of eltrombopag treatment. Bleeding decreased with eltrombopag treatment as compared with baseline. Eltrombopag was generally well tolerated; one patient experienced a transient ischemic attack on day 9. Eltrombopag (12.5-50 mg) is effective for the management of Japanese patients with chronic ITP (NCT00540423).


Subject(s)
Asian People , Benzoates/administration & dosage , Blood Platelets/drug effects , Hematologic Agents/administration & dosage , Hemorrhage/prevention & control , Hydrazines/administration & dosage , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Pyrazoles/administration & dosage , Administration, Oral , Adult , Aged , Benzoates/adverse effects , Benzoates/pharmacokinetics , Blood Platelets/immunology , Blood Platelets/metabolism , Chronic Disease , Double-Blind Method , Female , Hematologic Agents/adverse effects , Hematologic Agents/pharmacokinetics , Hemorrhage/blood , Hemorrhage/ethnology , Hemorrhage/immunology , Humans , Hydrazines/adverse effects , Hydrazines/pharmacokinetics , Japan/epidemiology , Male , Middle Aged , Placebos , Platelet Count , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/ethnology , Purpura, Thrombocytopenic, Idiopathic/immunology , Pyrazoles/adverse effects , Pyrazoles/pharmacokinetics , Receptors, Thrombopoietin/agonists , Receptors, Thrombopoietin/blood , Time Factors , Treatment Outcome
6.
Br J Biomed Sci ; 68(1): 19-22, 2011.
Article in English | MEDLINE | ID: mdl-21473257

ABSTRACT

Polymerase chain reaction (PCR) amplification was performed on 20 isolates of five Campylobacter species using a degenerate primer pair designed in silico to generate a product of the luxS gene or its homologue from Campylobacter organisms. Although the primer pair successfully amplified products of approximately 500 base pairs (bp) with the eight isolates of C. jejuni and C. coli and some of C. upsaliensis and C. fetus, it failed to amplify fragments with all four isolates of C. lari (two urease-negative C. lari; two urease-positive thermophilic campylobacters). When Southern blot hybridisation analysis was carried using the mixed luxS gene fragments prepared from the C. jejuni, C. coli, C. upsaliensis and C. fetus strains as a probe, all C. jejuni, C. coli, C. upsaliensis and C. fetus isolates gave positive signals, but no positive signal was detected with any C. lari isolate. These results clearly indicate that C. jejuni, C. coli, C. upsaliensis and C. fetus carry the luxS gene or its homologue. However, no luxS gene or its homologue was identified to occur in the C. lari genome. Although autoinducer-2 assays were positive in C. jejuni, C. coli, C. upsaliensis and C. fetus isolates, it was negative with all the C. lari isolates examined. In addition, a biofilm formation assay demonstrated that biofilm formation in the C. lari species does not appear to correlate with the occurrence of the luxS gene because biofilm formation occurred among some isolates of C. lari.


Subject(s)
Bacterial Proteins/genetics , Campylobacter/genetics , Carbon-Sulfur Lyases/genetics , DNA, Bacterial/genetics , Biofilms , Blotting, Southern , Campylobacter/isolation & purification , Homoserine/analogs & derivatives , Homoserine/genetics , Lactones , Molecular Sequence Data , Polymerase Chain Reaction
7.
J Thromb Haemost ; 9(5): 1040-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21342433

ABSTRACT

BACKGROUND: Thromboxane A(2) receptor (TXA(2)R) abnormality appears to dominantly disturb platelet function. OBJECTIVES: To reveal a molecular genetic defect in a patient with TXA(2)R abnormality and investigate the mechanism for the impaired response to TXA(2). PATIENT: The proband (OSP-2, PT) was a 7-year-old Japanese girl, suffering from repeated mucocutaneous bleeding. METHODS AND RESULTS: U46619 (2.5 and 10 µm)-induced platelet aggregation was remarkably impaired in the proband and her father. Immunoblots showed that TXA(2)R expression levels in their platelets were approximately 50% of controls, and nucleotide sequence analysis revealed that they were heterozygous for a novel mutation, c.167dupG in the TXA(2)R cDNA. Expression studies using Chinese hamster ovary (CHO) cells indicated that the mutation is responsible for the expression defect in TXA(2)R. We then examined α(IIb)ß(3) activation by employing an initial velocity analysis and revealed that U46619 failed to induce a sustained α(IIb)ß(3) and Rap1B activation in the proband. In addition, platelet secretion as monitored by P-selectin expression was markedly impaired in response to U46619 but not to ADP. The interaction between secreted ADP and P2Y(12) has been shown to play a critical role in the sustained α(IIb)ß(3) activation (Kamae et al. J Thromb Haemost 2006; 4: 1379). As expected, small amounts of exogenous ADP (0.5 µm) partially restored the sustained α(IIb)ß(3) activation induced by U46619. CONCLUSION: Our present data strongly suggest that the impaired platelet activation in response to U46619 in the heterozygous subject for the TXA(2)R mutation is, at least in part, as a result of the decrease in ADP secretion.


Subject(s)
Mutation , Receptors, Thromboxane A2, Prostaglandin H2/genetics , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Blood Platelets/metabolism , CHO Cells , Child , Cricetinae , Cricetulus , Female , Hemorrhage , Heterozygote , Humans , Male , Parents , Platelet Aggregation/drug effects , Platelet Function Tests , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, G-Protein-Coupled/genetics
8.
J Thromb Haemost ; 7(8): 1373-83, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19522742

ABSTRACT

BACKGROUND: Glycoprotein (GP)VI deficiency is a rare platelet disorder with a mild bleeding tendency. However, its pathophysiology remains unclear. OBJECTIVES: We characterized a novel GPVI-deficient patient with immune thrombocytopenic purpura and searched for the presence of anti-GPVI autoantibodies in this and another patient with GPVI deficiency. METHODS AND RESULTS: A 12-year-old Japanese girl (case 1) with moderate thrombocytopenia and mild bleeding showed selectively impaired collagen-induced platelet aggregation. Flow cytometric analysis indicated that the patient had a defect in the expression of GPVI-FcRgamma. An eluate of her platelet-associated IgG contained anti-alpha(IIb)beta3 autoantibodies. Moreover, using GPVI-FcRgamma-transfected cells, we unexpectedly identified anti-GPVI antibodies against the soluble ectodomain of GPVI in the eluate, despite the patient's GPVI deficiency. In contrast, anti-GPVI antibodies were not detectable in her plasma. In another case of GPVI deficiency (case 2) without detectable plasma anti-GPVI antibodies, we again detected platelet-associated anti-GPVI antibodies. In a 2-year follow-up of case 1, the platelet count increased to within the normal range and the bleeding tendency improved. Interestingly, GPVI was again expressed on her platelets, in association with a decrease in the relative amount of anti-GPVI antibodies. CONCLUSIONS: This is the first demonstration of platelet-associated anti-GPVI antibodies in GPVI-deficient subjects, in one case with spontaneous restoration of GPVI expression. These results strongly suggest an autoimmune mechanism in GPVI deficiency.


Subject(s)
Autoantibodies/analysis , Blood Platelets/immunology , Platelet Membrane Glycoproteins/immunology , Adult , Child , Epitopes , Female , Humans , Platelet Membrane Glycoproteins/biosynthesis , Platelet Membrane Glycoproteins/deficiency , Purpura, Thrombocytopenic, Idiopathic/immunology
9.
J Thromb Haemost ; 4(6): 1379-87, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16706985

ABSTRACT

OBJECTIVE: Platelet integrin alpha(IIb)beta3 plays a crucial role in platelet aggregation, and the affinity of alpha(IIb)beta3 for fibrinogen is dynamically regulated. Employing modified ligand-binding assays, we analyzed the mechanism by which alpha(IIb)beta3 maintains its high-affinity state. METHODS AND RESULTS: Washed platelets adjusted to 50 x 10(3) microL(-1) were stimulated with 0.2 U mL(-1) thrombin or 5 microm U46619 under static conditions. After the completion of alpha(IIb)beta3 activation and granule secretion, different kinds of antagonists were added to the activated platelets. The activated alpha(IIb)beta3 was then detected by fluorescein isothiocyanate (FITC)-labeled PAC1. The addition of 1 mum AR-C69931MX (a P2Y12 antagonist) or 1 mm A3P5P (a P2Y1 antagonist) disrupted the sustained alpha(IIb)beta3 activation by approximately 92% and approximately 38%, respectively, without inhibiting CD62P or CD63 expression. Dilution of the platelet preparation to 500 microL(-1) also disrupted the sustained alpha(IIb)beta3 activation, and the disruption by such dilution was abrogated by the addition of exogenous adenosine 5'-diphosphate (ADP) in a dose-dependent fashion. The amounts of ADP released from activated platelets determined by high-performance liquid chromatography were compatible with the amounts of exogenous ADP required for the restoration. We next examined the effects of antagonists on protein kinase C (PKC) and Rap1B activation induced by 0.2 U mL(-1) thrombin. Thrombin induced long-lasting PKC and Rap1B activation. AR-C69931MX markedly inhibited Rap1B activation without inhibiting PKC activation. CONCLUSIONS: Our data indicate that the continuous interaction between released ADP and P2Y12 is critical for the maintenance of alpha(IIb)beta3 activation.


Subject(s)
Adenosine Diphosphate/metabolism , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Purinergic P2/metabolism , rap GTP-Binding Proteins/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Antibodies, Monoclonal , Blood Platelets/drug effects , Blood Platelets/enzymology , Dose-Response Relationship, Drug , Humans , Protein Kinase C/metabolism , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/immunology , Receptors, Purinergic P2Y12 , Signal Transduction , Thrombin/pharmacology
10.
J Thromb Haemost ; 3(10): 2315-23, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16194207

ABSTRACT

In this study, we have identified a patient (OSP-1) with a congenital P2Y12 deficiency showing a mild bleeding tendency from her childhood and examined the role of P2Y12 in platelet function. At low concentrations of agonists OSP-1 platelets showed an impaired aggregation to several kinds of stimuli, whereas at high concentrations they showed a specifically impaired platelet aggregation to adenosine diphosphate (ADP). ADP normally induced platelet shape change and failed to inhibit PGE1-stimulated cAMP accumulation in OSP-1 platelets. Molecular genetic analysis revealed that OSP-1 was a homozygous for a mutation in the translation initiation codon (ATG to AGG) in the P2Y12 gene. Heterologous cell expression of wild-type or mutant P2Y12 confirmed that the mutation was responsible for the deficiency in P2Y12. OSP-1 platelets showed a markedly impaired platelet spreading onto immobilized fibrinogen. Real-time observations of thrombogenesis under a high shear rate (2000 s(-1)) revealed that thrombi over collagen were small and loosely packed and most of the aggregates were unable to resist against high shear stress in OSP-1. Our data suggest that secretion of endogenous ADP and subsequent P2Y12-mediated signaling are critical for platelet aggregation, platelet spreading, and as a consequence, for stabilization of thrombus.


Subject(s)
Blood Platelet Disorders/genetics , Blood Platelets/pathology , Codon, Initiator/genetics , Membrane Proteins/deficiency , Point Mutation , Receptors, Purinergic P2/deficiency , Aged , Cell Line , Cell Shape/genetics , Cyclic AMP/analysis , DNA Mutational Analysis , Female , Homozygote , Humans , Membrane Proteins/genetics , Platelet Aggregation/genetics , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y12 , Thrombosis/genetics , Transfection
11.
J Thromb Haemost ; 3(10): 2307-14, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16150045

ABSTRACT

BACKGROUND: The alphaIIbbeta3 antagonists inhibit platelet aggregation and are used as antithrombotic agents for cardiothrombotic disease. The present study investigates the correlation of inhibition of fibrinogen and von Willebrand factor (VWF) binding by alphaIIbbeta3 antagonists with the inhibition of platelet aggregation and prolongation of bleeding time (BT). METHODS: Inhibition of fibrinogen and VWF binding were assessed in a purified alphaIIbbeta3-binding assay. As an in vitro cell-based assay, platelet aggregation and VWF-mediated adhesion studies were performed using human platelets. In vivo effects on BT were measured using a template device in dogs at the same time as an ex vivo aggregation study was performed. RESULTS: In vitro studies demonstrated that the antiaggregatory effects of alphaIIbbeta3 antagonists correlate with their inhibition of fibrinogen binding, but not VWF. Interestingly, the effects of alphaIIbbeta3 antagonists on BT could be differentiated from the inhibition of platelet aggregation. Furthermore, this differentiation was strongly correlated with the different inhibitory potencies between fibrinogen and VWF binding, as well as that between VWF-mediated adhesion and aggregation. CONCLUSIONS: Our study provides novel evidence showing that the inhibitory effect of alphaIIbbeta3 antagonists on VWF, but not fibrinogen binding, correlates with their ability to prolong BT.


Subject(s)
Blood Platelets/physiology , Fibrinogen/antagonists & inhibitors , Hemorrhage/etiology , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , von Willebrand Factor/antagonists & inhibitors , Animals , Blood Coagulation Tests , Dogs , Fibrinogen/metabolism , Humans , Kinetics , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Protein Binding/drug effects , von Willebrand Factor/metabolism
12.
J Thromb Haemost ; 3(2): 361-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15670045

ABSTRACT

WAVE isoforms, which consist of WAVE-1, WAVE-2 and WAVE-3, are members of the Wiskott-Aldrich syndrome protein (WASP) family. They are implicated in the regulation of actin-cytoskeletal reorganization downsteam of the small GTPase, Rac. Since platelet attachment to extracellular matrices leads to filopodial and lamellipodial extension, we examined the expression and subcellular localization of WAVEs in platelets. Employing primary megakaryocytic cells derived from cord blood as well as megakaryocytic cell lines, we also examined their expression during megakaryocytic differentiation. Immunoblotting and immunohistochemical analysis revealed that platelets expressed WAVE-1 and WAVE-2, whereas WAVE-3 expression was hardly to be detected. WAVE-1 expression was associated with megakaryocytic differentiation, whereas WAVE-2 and WAVE-3 expression was not changed by the differentiation. In adhered platelets both WAVE-1 and WAVE-2 were localized at the edge of the lamellipodia and at the tips of filopodia. In WASP-deficient platelets we found normal lamellipodial formation and localization of WAVE-1 and WAVE-2 at the edges of lamellipodia. Furthermore, we demonstrated that WAVE-1 and WAVE-2 moved from a detergent-soluble cytosolic fraction to insoluble cytoskeleton fraction after platelet aggregation. These results suggest that WAVE-1 and WAVE-2 regulate actin reorganization during platelet spreading and aggregate formation.


Subject(s)
Blood Platelets/chemistry , Megakaryocytes/chemistry , Microfilament Proteins/metabolism , Blood Platelets/cytology , Cell Differentiation , Cell Lineage , Cytoskeleton/metabolism , Cytosol/metabolism , Humans , Megakaryocytes/cytology , Microfilament Proteins/analysis , Platelet Adhesiveness , Protein Transport , Proteins , Pseudopodia/chemistry , Wiskott-Aldrich Syndrome Protein Family
13.
J Thromb Haemost ; 2(1): 177-86, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14717982

ABSTRACT

Affinity/avidity state of integrin alpha IIb beta 3 is regulated by intracellular inside-out signaling. Although several megakaryocytic cell lines have been established, soluble ligand binding to alpha IIb beta 3 expressed in these cells by cellular agonists has not been demonstrated. We have re-examined agonist-induced alpha IIb beta 3 activation on megakaryocytic cell lines with a marker of the late stage of megakaryocytic differentiation, glycoprotein Ib (GPIb). Activation of alpha IIb beta 3 was assessed by PAC1 and soluble fibrinogen binding to the cells. We found that alpha IIb beta 3 expressed in CMK cells with high GPIb expression was activated by a phorbor ester, phorbol myristate acetate (PMA). Although the population of the GPIbhigh cells was <0.5% of the total cells, incubation with a nucleoside analog, ribavirin, efficiently increased the PMA-reactive GPIbhigh cells. Not only PMA but also a calcium ionophore, A23187, induced alpha IIb beta 3 activation, and PMA and A23187 had an additive effect on alpha IIb beta 3 activation. Ligand binding to the activated alpha IIb beta 3 in the GPIbhigh CMK cells is totally abolished by an alpha IIb beta 3-specific antagonist, and inhibited by wortmannin, cytochalasin-D and prostaglandin E1, and the effects of these inhibitors on alpha IIb beta 3 activation in the GPIbhigh CMK cells were compatible with those in platelets. We have also demonstrated that the ribavirin-treated CMK cells express PKC-alpha, -beta, -delta and -theta, and suggested that PKC-alpha and/or -beta appear to be responsible for PMA-induced activation of alpha IIb beta 3 in CMK cells.


Subject(s)
Megakaryocytes/immunology , Megakaryocytes/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Alprostadil/pharmacology , Androstadienes/pharmacology , Calcimycin/pharmacology , Cell Line , Cytochalasin D/pharmacology , Humans , Ionophores/pharmacology , Isoenzymes/metabolism , Megakaryocytes/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/agonists , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Protein Kinase C/metabolism , Ribavirin/pharmacology , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Wortmannin
14.
Bone Marrow Transplant ; 31(11): 1057-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12774060

ABSTRACT

We report the successful treatment of two acute lympho- blastic leukemia (ALL) patients who relapsed following allogeneic bone marrow transplantation (allo-BMT) with allogeneic peripheral blood sem cell transplantation(allo-PBSCT) and donor lymphocyte infusion (DLI) from the same HLA-identical related donors as those used for the first allo-BMT. The patients relapsed on days 154 and 351 from the initial allo-BMT, respectively. Since conventional reinduction chemotherapy failed, allo-PBSCT was undertaken while the patients were still myelosuppressed immediately after reinduction chemotherapy. To induce and/or enhance GVL effects following allo-PBSCT, we performed rapid tapering of CsA and added DLI. After allo-PBSCT and DLI, the patients maintained their complete remission at 55 and 48 months post allo-PBSCT, respectively. From these findings, allo-PBSCT and DLI may be a useful treatment strategy for acute leukemia relapsing after allo-BMT.


Subject(s)
Lymphocyte Transfusion , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Stem Cell Transplantation/methods , Adult , Antineoplastic Agents/therapeutic use , Bone Marrow Transplantation , Female , Graft vs Host Disease/prevention & control , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/surgery , Transplantation Conditioning/methods , Treatment Outcome
15.
Ann Hematol ; 81(4): 224-7, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11976826

ABSTRACT

We experienced a rare case of a lymphomatous polyp of mantle cell type forming a polypoid mass lesion in the duodenum bulbous together with advanced gastric cancer. A total gastrectomy was performed, and the specimen revealed atypical small- to medium-sized lymphoid cells with indented nuclei, which infiltrated the Peyer's patch and formed a nodular mass in the lamina propria and submucosa of the duodenum. The lymphoma cells also infiltrated the lymphoid follicle of the gastric mucosa, spleen, and regional lymph node with a typical mantle zone pattern. Flow cytometric analysis of the single cells of the lymph node and immunohistochemistry of a paraffin-embedded specimen revealed that the lymphoma cells expressed surface CD5, CD19, CD20, and nuclear cyclin D1. Chromosomal analysis of this single cell suspension revealed that these lymphoma cells have trisomy 3 in conjunction with t(11;14)(q13;q32), which is frequently seen in mucosa-associated lymphoid tissue lymphomas (MALToma) in the stomach and is also reported in mantle cell lymphoma as a secondary genetic alteration. Our report suggests that trisomy 3 may be a common chromosomal abnormality in lymphomatous polyps of mantle cell type.


Subject(s)
Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 14 , Duodenal Neoplasms/pathology , Intestinal Polyps/pathology , Lymphoma, Mantle-Cell/pathology , Stomach Neoplasms/pathology , Translocation, Genetic , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Chromosomes, Human, Pair 3 , Clone Cells/immunology , Clone Cells/pathology , Cytogenetic Analysis , Duodenal Neoplasms/diagnosis , Duodenal Neoplasms/genetics , Humans , Immunophenotyping , Intestinal Polyps/diagnosis , Intestinal Polyps/genetics , Lymphoma, Mantle-Cell/diagnosis , Lymphoma, Mantle-Cell/genetics , Male , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasms, Second Primary/diagnosis , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/pathology , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Trisomy
16.
J Pediatr Surg ; 36(12): 1838-40, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733919

ABSTRACT

The authors experienced an extremely rare case of secondary sclerosing cholangitis and portal hypertension developed as late complications of hemolytic uremic syndrome (HUS) owing to Escherichia coli O157:H7 in a 2-year-old boy. HUS after E coli O157 infection is the most frequent cause of acute renal failure in childhood and occasionally is accompanied by extrarenal complications such as encephalopathy, cardiomyopathy, ischemic colitis, and pancreatitis. Rarely, late colonic stenosis may develop secondary to the ischemic damage. Sclerosing cholangitis and subsequent cirrhosis with portal hypertension are very uncommon as late complications of HUS. To our knowledge, such a case has not been previously reported in the literature. J Pediatr Surg 36:1838-1840.


Subject(s)
Cholangitis, Sclerosing/etiology , Enterocolitis/complications , Escherichia coli Infections/complications , Hemolytic-Uremic Syndrome/complications , Hypertension, Portal/etiology , Child, Preschool , Escherichia coli O157/chemistry , Humans , Male , Shiga Toxins/adverse effects
17.
J Histochem Cytochem ; 49(11): 1397-405, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11668193

ABSTRACT

Two mannose 6-phosphate receptors, cation-dependent and -independent receptors (CDMPR and CIMPR), play an important role in the intracellular transport of lysosomal enzymes. To investigate functional differences between the two in vivo, their distribution was examined in the rat liver using immunohistochemical techniques. Positive signals corresponding to CIMPR were detected intensely in hepatocytes and weakly in sinusoidal Kupffer cells and interstitial cells in Glisson's capsule. In the liver acinus, hepatocytes in the perivenous region showed a more intense immunoreactivity than those in the periportal region. On the other hand, positive staining of CDMPR was detected at a high level in Kupffer cells, epithelial cells of interlobular bile ducts, and fibroblast-like cells, but the corresponding signal was rather weak in hepatocytes. In situ hybridization analysis also revealed a high level of expression of CIMPR mRNAs in hepatocytes and of CDMPR mRNA in Kupffer cells. By double immunostaining, OX6-positive antigen-presenting cells in Glisson's capsule were co-labeled with the CDMPR signal but were only faintly stained with anti-CIMPR. These different distribution patterns of the two MPRs suggest distinct functional properties of each receptor in liver tissue.


Subject(s)
Liver/metabolism , Receptor, IGF Type 2/metabolism , Animals , Antibody Specificity , Cations , Immunoblotting , In Situ Hybridization , Microscopy, Fluorescence , Rats , Rats, Wistar
18.
Blood ; 98(6): 1819-27, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11535516

ABSTRACT

Localization of epitopes for platelet-associated (PA) anti-GPIIb-IIIa (alpha(IIb)beta(3)) autoantibodies in chronic immune thrombocytopenic purpura remains elusive. Previous studies suggest that PA antibodies recognize the tertiary structure of intact glycoprotein (GP) IIb-IIIa. To localize their epitopes using antigen-capture enzyme-linked immunosorbent assay (ELISA), the reactivity of 34 PA anti-GPIIb-IIIa antibodies was examined with recombinant GPIIb-IIIa having a defect in ligand-binding sites in either GPIIb or GPIIIa, and no major conformational change was induced: KO variant GPIIb-IIIa was attributed to a 2-amino acid insertion between residues 160 and 161 in the W3 4-1 loop in GPIIb, and CAM variant GPIIb-IIIa was attributed to D119Y in GPIIIa. In one third (11 of 34) of the patients, PA antibodies showed a marked decrease (less than 50%) in reactivity with KO compared with wild-type GPIIb-IIIa. Their reactivity was also impaired against GPIIbD163A-IIIa. In sharp contrast, they reacted normally with CAM GPIIb-IIIa. OP-G2, a ligand-mimetic monoclonal antibody, markedly inhibited their binding to GPIIb-IIIa in patients with impaired binding to KO GPIIb-IIIa, but small GPIIb-IIIa antagonists did not. In addition, a newly developed sensitive ELISA indicated that autoantibodies showing impaired binding to KO are more potent inhibitors for fibrinogen binding. The present data suggest that certain PA anti-GPIIb-IIIa autoantibodies recognize epitopes close to the ligand-binding site in GPIIb, but not in GPIIIa.


Subject(s)
Autoantibodies/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Adult , Aged , Antibodies, Monoclonal/immunology , Autoantigens/genetics , Autoantigens/immunology , Binding Sites , Blood Platelets/immunology , Cell Line , Chronic Disease , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Female , Fibrinogen/metabolism , Humans , Male , Middle Aged , Mutation , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
19.
Cytokine Growth Factor Rev ; 12(4): 337-48, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11544103

ABSTRACT

The type I interferon (IFN) family includes IFN-alpha, IFN-beta, IFN-pi, and IFN-tau. These molecules are clustered according to sequence homologies, use of the same cell surface receptor, and similar functions. IFN-alpha and IFN-beta have a globular structure composed of five a-helices. Their receptors, IFNAR1 and IFNAR2, belong to the class II cytokine receptor family for a-helical cytokines. Information about structure-function relationships between these and other IFNs is being provided by comparative sequence analysis, reference to a prototypic three-dimensional structure, analysis with monoclonal antibodies, construction of hybrid molecules and site directed mutagenesis. While much remains to be done, it should someday be possible to understand differences among IFNs in terms of how they interact with their corresponding receptors. Our recently identified IFN-like molecule, limitin, has weak sequence homology to IFN-alpha, IFN-beta, and IFN-omega and displays its biological functions through the same IFN-alpha/beta receptors. While limitin has antiproliferative, immunomodulatory, and antiviral effects like IFN-alpha and IFN-beta, it is unique in lacking influence on myeloid and erythroid progenitors. Further analysis of this functionally unique cytokine should be informative about complex IFN-receptor interactions. Furthermore, a human homologue or synthetic variant might be superior for clinical applications as an IFN without myelosuppressive properties.


Subject(s)
Cytokines/chemistry , Cytokines/physiology , Interferon Type I/chemistry , Interferon Type I/physiology , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data
20.
J Immunol ; 167(6): 3156-63, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11544301

ABSTRACT

A novel IFN-like molecule, limitin, was recently identified and revealed to suppress B lymphopoiesis through the IFN-alphabeta receptor, although it lacked growth suppression on myeloid and erythroid progenitors. Here we have studied diverse effects of limitin on T lymphocytes and compared limitin with previously known IFNs. Like IFN-alpha and -beta, limitin modified immunity in the following responses. It suppressed mitogen- and Ag-induced T cell proliferation through inhibiting the responsiveness to exogenous IL-2 rather than suppressing the production of IL-2. In contrast, limitin enhanced cytotoxic T lymphocyte activity associated with the perforin-granzyme pathway. To evaluate the effect of limitin in vivo, a lethal graft-versus-host disease assay was established. Limitin-treatment of host mice resulted in the enhancement of graft-versus-host disease. Limitin did not influence thymocyte development either in fetal thymus organ cultures or in newborn mice injected with limitin-Ig, suggesting that limitin is distinguishable from IFN-alpha and -beta. From these findings, it can be speculated that the human homolog of limitin may be applicable for clinical usage because of its IFN-like activities with low adverse effects on, for example, T lymphopoiesis, erythropoiesis, and myelopoiesis.


Subject(s)
Cytokines/physiology , Lymphocyte Activation/drug effects , T-Lymphocyte Subsets/drug effects , Adjuvants, Immunologic/pharmacology , Animals , Cell Division/drug effects , Cytokines/drug effects , Cytokines/toxicity , Cytotoxicity, Immunologic , Drug Evaluation, Preclinical , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Hematopoiesis/drug effects , Immunosuppressive Agents/pharmacology , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Interleukin-2/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Muromonab-CD3/pharmacology , Organ Culture Techniques , Ovalbumin/immunology , Radiation Chimera , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...