Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Blood ; 140(9): 1038-1051, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35767715

ABSTRACT

Protein tyrosine phosphatase nonreceptor type 22 (PTPN22) is a protein tyrosine phosphatase that negatively regulates T-cell signaling. However, whether it is expressed and functions in platelets remains unknown. Here we investigated the expression and role of PTPN22 in platelet function. We reported PTPN22 expression in both human and mouse platelets. Using PTPN22-/- mice, we showed that PTPN22 deficiency significantly shortened tail-bleeding time and accelerated arterial thrombus formation without affecting venous thrombosis and the coagulation factors VIII and IX. Consistently, PTPN22-deficient platelets exhibited enhanced platelet aggregation, granule secretion, calcium mobilization, lamellipodia formation, spreading, and clot retraction. Quantitative phosphoproteomic analysis revealed the significant difference of phosphodiesterase 5A (PDE5A) phosphorylation in PTPN22-deficient platelets compared with wild-type platelets after collagen-related peptide stimulation, which was confirmed by increased PDE5A phosphorylation (Ser92) in collagen-related peptide-treated PTPN22-deficient platelets, concomitant with reduced level and vasodilator-stimulated phosphoprotein phosphorylation (Ser157/239). In addition, PTPN22 interacted with phosphorylated PDE5A (Ser92) and dephosphorylated it in activated platelets. Moreover, purified PTPN22 but not the mutant form (C227S) possesses intrinsic serine phosphatase activity. Furthermore, inhibition of PTPN22 enhanced human platelet aggregation, spreading, clot retraction, and increased PDE5A phosphorylation (Ser92). In conclusion, our study shows a novel role of PTPN22 in platelet function and arterial thrombosis, identifying new potential targets for future prevention of thrombotic or cardiovascular diseases.


Subject(s)
Hemostasis , Protein Tyrosine Phosphatase, Non-Receptor Type 22 , Thrombosis , Animals , Blood Platelets/metabolism , Humans , Mice , Mice, Knockout , Platelet Activation , Platelet Aggregation , Platelet Function Tests , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Thrombosis/genetics
2.
Ann Hematol ; 99(10): 2315-2322, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32728937

ABSTRACT

Immune thrombocytopenia (ITP) is an autoimmune disease characterized by lower platelet count resulting from immune cells-mediated platelet clearance. Tacrolimus is an immunosuppressive agent which selectively inhibits T cell activation. Whether tacrolimus plays a role in ITP remains unclear. This study aimed to investigate the effect of tacrolimus on ITP in mice. An ITP mouse model was established by injection of rat anti-mouse integrin GPIIb/CD41 immunoglobulin and treated with tacrolimus followed by isolation of peripheral blood mononuclear cells and plasma. The mRNA expression of T-bet, GATA3, and Foxp3 was measured by RT-PCR, and level of IFN-γ, IL-12p70, IL-4, IL-13, and TGF-ß in plasma was measured by ELISA. Tacrolimus inhibited antiplatelet antibody-mediated platelet clearance in ITP mouse model. Meanwhile, tacrolimus-treated ITP mice displayed a significant decrease in the mRNA expression of T-bet and plasma level of IFN-γ and IL-12p70 compared with ITP mice but without differences when compared with normal mice. Furthermore, the expression of GATA3, Foxp3, and plasma level of IL-4 and TGF-ß were upregulated in tacrolimus-treated ITP mice without significant differences to normal mice (except TGF-ß). Tacrolimus prevents antiplatelet antibody-mediated thrombocytopenia in ITP mice possibly through regulating T cell differentiations, suggesting it might be a novel approach for preventing ITP.


Subject(s)
Immunosuppressive Agents/therapeutic use , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Tacrolimus/therapeutic use , Animals , Blood Platelets/immunology , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Isoantibodies/blood , Mice , Mice, Inbred C57BL , Purpura, Thrombocytopenic, Idiopathic/genetics , Purpura, Thrombocytopenic, Idiopathic/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Transcription Factors/biosynthesis , Transcription Factors/genetics
3.
Appl Opt ; 58(36): 9734-9739, 2019 Dec 20.
Article in English | MEDLINE | ID: mdl-31873613

ABSTRACT

The separation of misalignment aberrations is a crucial step in interferometric testing for the acquisition of the real surface. In this paper, a Fizeau interference system and a conical mirror with a certain angle were used to achieve the shape measurement of the inner surface of the frustum of a cone. Based on the ray-tracing method, the relationship between the adjustment errors and misalignment aberrations was established, and the misalignment aberrations could be removed by using the error separation model that accords with the least-square algorithm theory. The simulation and practical measurement results indicate that the approach proposed is valid and feasible.

4.
J Transl Med ; 16(1): 311, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30442147

ABSTRACT

BACKGROUND: Platycodin D (PD) is one of the major bioactive components of the roots of Platycodon grandiflorum and possesses multiple biological and pharmacological properties, such as antiviral, anti-inflammatory, and anti-cancer activities. However, whether it affects platelet function remains unclear. This study aims to evaluate the role of PD in platelet function and thrombus formation. METHODS: Platelets were treated with PD followed by measuring platelet aggregation, activation, spreading, clot retraction, expression of glycoprotein receptors. Moreover, mice platelets were treated with PD and infused into wild-type mice for analysis of in vivo hemostasis and arterial thrombosis. RESULTS: Platycodin D treatment significantly inhibited platelet aggregation in response to collagen, ADP, arachidonic acid and epinephrine, reduced platelet P-selectin expression, integrin αIIbß3 activation, spreading on fibrinogen as well as clot retraction, accompanied with decreased phosphorylation of Syk and PLCγ2 in collagen-related peptide or thrombin-stimulated platelets. Moreover, PD-treated mice platelets presented significantly impaired in vivo hemostasis and arterial thrombus formation. Interestingly, PD induced internalization of glycoprotein receptors αIIbß3, GPIbα and GPVI. However, GM6001, cytochalasin D, BAPTA-AM and wortmannin did not prevent PD-induced internalization of receptors. CONCLUSIONS: Our study demonstrates that PD inhibits platelet aggregation, activation and impairs hemostasis and arterial thrombosis, suggesting it might be a potent anti-thrombotic drug.


Subject(s)
Blood Platelets/metabolism , Blood Platelets/pathology , Endocytosis/drug effects , Platelet Membrane Glycoproteins/metabolism , Saponins/pharmacology , Thrombosis/pathology , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Arteries/drug effects , Arteries/pathology , Blood Platelets/drug effects , Clot Retraction/drug effects , Hemostasis/drug effects , Humans , Mice, Inbred C57BL , P-Selectin/metabolism , Phospholipase C gamma/metabolism , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Signal Transduction/drug effects , Syk Kinase/metabolism
5.
Haematologica ; 103(9): 1568-1576, 2018 09.
Article in English | MEDLINE | ID: mdl-29794149

ABSTRACT

In addition to their hemostatic function, platelets play an important role in regulating the inflammatory response. The platelet NLRP3 inflammasome not only promotes interleukin-1ß secretion, but was also found to be upregulated during platelet activation and thrombus formation in vitro However, the role of NLRP3 in platelet function and thrombus formation in vivo remains unclear. In this study, we aimed to investigate the role of NLRP3 in platelet integrin αIIbß3 signaling transduction. Using NLRP3-/- mice, we showed that NLRP3-deficient platelets do not have significant differences in expression of the platelet-specific adhesive receptors αIIbß3 integrin, GPIba or GPVI; however, NLRP3-/- platelets transfused into wild-type mice resulted in prolonged tail-bleeding time and delayed arterial thrombus formation, as well as exhibiting impaired spreading on immobilized fibrinogen and defective clot retraction, concomitant with decreased phosphorylation of c-Src, Syk and PLCγ2 in response to thrombin stimulation. Interestingly, addition of exogenous recombinant interleukin-1ß reversed the defect in NLRP3-/- platelet spreading and clot retraction, and restored thrombin-induced phosphorylation of c-Src/Syk/PLCγ2, whereas an anti-interleukin-1ß antibody blocked spreading and clot retraction mediated by wild-type platelets. Using the direct NLRP3 inhibitor, CY-09, we demonstrated significantly reduced human platelet aggregation in response to threshold concentrations of collagen and ADP, as well as impaired clot retraction in CY-09-treated human platelets, supporting a role for NLRP3 also in regulating human platelet αIIbß3 outside-in signaling. This study identifies a novel role for NLRP3 and interleukin-1ß in platelet function, and provides a new potential link between thrombosis and inflammation, suggesting that therapies targeting NLRP3 or interleukin-1ß might be beneficial for treating inflammation-associated thrombosis.


Subject(s)
Blood Platelets/metabolism , Hemostasis , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Signal Transduction , Thrombosis/etiology , Thrombosis/metabolism , Animals , Biomarkers , Blood Coagulation/genetics , Cell Degranulation , Clot Retraction , Disease Models, Animal , Gene Expression , Humans , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Platelet Activation , Platelet Aggregation/genetics , Platelet Function Tests , Thrombosis/pathology
6.
Zhonghua Zhong Liu Za Zhi ; 37(3): 213-5, 2015 Mar.
Article in Zh | MEDLINE | ID: mdl-25975792

ABSTRACT

OBJECTIVE: To find a rational way in early detecting the residual tumor in patients with nasopharyngeal carcinoma (NPC) after radiotherapy. METHODS: A total of 47 NPC patients who were diagnosed residual tumor after radiotherapy and treated in our hospital from Dec 2009 to Aug 2012 were included in this study and their clinicopathological and follow-up data were reviewed and analyzed. The patients were checked by nasopharynx MRI, FDG PET-CT and were examined by biopsy of the residual tumors within two weeks after radiotherapy. The diagnosis of relapses was determined by pathological re-examination. RESULTS: All the 47 patients were followed up for 10-42 months. Three of them had nasopharynx relapse. The others had not tumor relapse and their residual tumors disappeared completely. The specificity of MRI, FDG PET-CT and pathological tumor response in diagnosing residual tumors were 9.1%, 77.3%, and 95.5% (P<0.001). Their accuracy rates were 14.9%, 78.9%, and 95.7%, respectively (P<0.001). The M of SUVmax in the team who had moderate and severe pathologic tumor response (team A) was 3.05 and that in the team who had mild pathologic tumor response (team B) was 4.68 (P=0.012). None of patients in the team A had nasopharynx relapse. Three patients in the team B who had SUVmax ≥4 had nasopharynx relapses, and in other 2 patients who had SUVmax <4, the residual tumors disappeared during the following-up. The specificity and accuracy in diagnosing residual tumors were increased when mild pathologic tumor response combined with SUVmax4 were used. CONCLUSION: PET-CT combined with pathologic tumor response is beneficial for early diagnosis of residual nasopharyngeal tumors after radiotherapy.


Subject(s)
Nasopharyngeal Neoplasms/diagnosis , Neoplasm, Residual/diagnosis , Positron-Emission Tomography , Tomography, X-Ray Computed , Carcinoma , Fluorodeoxyglucose F18 , Humans , Magnetic Resonance Imaging , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/diagnostic imaging , Nasopharyngeal Neoplasms/radiotherapy , Neoplasm Recurrence, Local , Neoplasm, Residual/diagnostic imaging , Neoplasm, Residual/radiotherapy , Sensitivity and Specificity
7.
Pol J Microbiol ; 73(1): 29-38, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38437465

ABSTRACT

Fungal diseases form perforated disease spots in tobacco plants, resulting in a decline in tobacco yield and quality. The present study investigated the antagonistic effect of Bacillus subtilis CTXW 7-6-2 against Rhizoctonia solani, its ability to promote the growth of tobacco seedlings, and the expression of disease resistance-related genes for efficient and eco-friendly plant disease control. Our results showed that CTXW 7-6-2 had the most vigorous growth after being cultured for 96 h, and its rate of inhibition of R. solani growth in vitro was 94.02%. The volatile compounds produced by CTXW 7-6-2 inhibited the growth of R. solani significantly (by 96.62%). The fungal growthinhibition rate of the B. subtilis CTXW 7-6-2 broth obtained after high-temperature and no-high-temperature sterile fermentation was low, at 50.88% and 54.63%, respectively. The lipopeptides extracted from the B. subtilis CTXW 7-6-2 fermentation broth showed a 74.88% fungal growth inhibition rate at a concentration of 100 mg/l. Scanning and transmission electron microscopy showed some organelle structural abnormalities, collapse, shrinkage, blurring, and dissolution in the R. solani mycelia. In addition, CTXW 7-6-2 increased tobacco seedling growth and improved leaf and root weight compared to the control. After CTXW 7-6-2 inoculation, tobacco leaves showed the upregulation of the PDF1.2, PPO, and PAL genes, which are closely related to target spot disease resistance. In conclusion, B. subtilis CTXW 7-6-2 may be an efficient biological control agent in tobacco agriculture and enhance plant growth potential.


Subject(s)
Bacillus subtilis , Nicotiana , Bacillus subtilis/genetics , Disease Resistance , Rhizoctonia
8.
Aging (Albany NY) ; 12(9): 8151-8166, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32352928

ABSTRACT

Salidroside is the main bioactive component in Rhodiola rosea and possesses multiple biological and pharmacological properties. However, whether salidroside affects platelet function remains unclear. Our study aims to investigate salidroside's effect on platelet function. Human or mouse platelets were treated with salidroside (0-20 µM) for 1 hour at 37°C. Platelet aggregation, granule secretion, and receptors expression were measured together with detection of platelet spreading and clot retraction. In addition, salidroside (20 mg/kg) was intraperitoneally injected into mice followed by measuring tail bleeding time, arterial and venous thrombosis. Salidroside inhibited thrombin- or CRP-induced platelet aggregation and ATP release and did not affect the expression of P-selectin, glycoprotein (GP) Ibα, GPVI and αIIbß3. Salidroside-treated platelets presented decreased spreading on fibrinogen or collagen and reduced clot retraction with decreased phosphorylation of c-Src, Syk and PLCγ2. Additionally, salidroside significantly impaired hemostasis, arterial and venous thrombus formation in mice. Moreover, in thrombin-stimulated platelets, salidroside inhibited phosphorylation of AKT (T308/S473) and GSK3ß (Ser9). Further, addition of GSK3ß inhibitor reversed the inhibitory effect of salidroside on platelet aggregation and clot retraction. In conclusion, salidroside inhibits platelet function and thrombosis via AKT/GSK3ß signaling, suggesting that salidroside may be a novel therapeutic drug for treating thrombotic or cardiovascular diseases.


Subject(s)
Blood Platelets/drug effects , Gene Expression Regulation , Glucosides/pharmacology , Glycogen Synthase Kinase 3 beta/genetics , Phenols/pharmacology , Proto-Oncogene Proteins c-akt/genetics , RNA/genetics , Thrombosis/prevention & control , Animals , Glycogen Synthase Kinase 3 beta/biosynthesis , Humans , Mice , Phosphorylation/drug effects , Platelet Activation/drug effects , Proto-Oncogene Proteins c-akt/biosynthesis , Rhodiola , Signal Transduction , Thrombosis/blood , Thrombosis/genetics
9.
Redox Biol ; 34: 101569, 2020 07.
Article in English | MEDLINE | ID: mdl-32422541

ABSTRACT

NADPH oxidase-derived reactive oxygen species (ROS) regulates platelet function and thrombosis. It remains controversial regarding NOX2's role in platelet function. As a regulatory subunit for NOX2, whether p47phox regulates platelet function remains unclear. Our study intends to evaluate p47phox's role in platelet function. Platelets were isolated from wild-type or p47phox-/- mice followed by analysis of platelet aggregation, granule secretion, surface receptors expression, spreading, clot retraction and ROS generation. Additionally, in vivo hemostasis, arterial and venous thrombosis was assessed. Moreover, human platelets were treated with PR-39 to inhibit p47phox activity followed by analysis of platelet function. p47phox deficiency significantly prolonged tail-bleeding time, delayed arterial and venous thrombus formation in vivo as well as reduced platelet aggregation, ATP release and αIIbß3 activation. In addition, p47phox-/- platelets presented impaired spreading on fibrinogen or collagen and defective clot retraction concomitant with decreased phosphorylation of Syk and PLCγ2. Moreover, CRP or thrombin-stimulated p47phox-/- platelets displayed reduced intracellular ROS generation which was further decreased after inhibition of NOX1. Meanwhile, p47phox deficiency increased VASP phosphorylation and decreased phosphorylation of ERK1/2, p38, ERK5 and JNK without affecting AKT and c-PLA2 phosphorylation. Furthermore, p47phox translocates to membrane to interact with both NOX1 and NOX2 after stimulation with CRP or thrombin. Finally, inhibition of p47phox activity by PR-39 reduced ROS generation, platelet aggregation and clot retraction in human platelets. In conclusion, p47phox regulates platelet function, arterial and venous thrombus formation and ROS generation, indicating that p47phox might be a novel therapeutic target for treating thrombotic or cardiovascular diseases.


Subject(s)
Platelet Activation , Venous Thrombosis , Animals , Blood Platelets , Hemostasis , Mice , NADPH Oxidases/genetics , Platelet Aggregation , Venous Thrombosis/genetics
10.
J Int Med Res ; 47(4): 1731-1739, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30799665

ABSTRACT

OBJECTIVE: To investigate the role of alteplase, a widely-used thrombolytic drug, in platelet function. METHODS: Human platelets were incubated with different concentrations of alteplase followed by analysis of platelet aggregation in response to adenosine diphosphate (ADP), collagen, ristocetin, arachidonic acid or epinephrine using light transmittance aggregometry. Platelet activation and surface levels of platelet receptors GPIbα, GPVI and αIIbß3 were analysed using flow cytometry. The effect of alteplase on clot retraction was also examined. RESULTS: This study demonstrated that alteplase significantly inhibited platelet aggregation in response to ADP, collagen and epinephrine in a dose-dependent manner, but it did not affect ristocetin- or arachidonic acid-induced platelet aggregation. Alteplase did not affect platelet activation as demonstrated by no differences in P-selectin levels and PAC-1 binding being observed in collagen-stimulated platelets after alteplase treatment compared with vehicle. There were no changes in the surface levels of the platelet receptors GPIbα, GPVI and αIIbß3 in alteplase-treated platelets. Alteplase treatment reduced thrombin-mediated clot retraction. CONCLUSIONS: Alteplase inhibits platelet aggregation and clot retraction without affecting platelet activation and surface receptor levels.


Subject(s)
Blood Platelets/physiology , Platelet Activation , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Tissue Plasminogen Activator/administration & dosage , Blood Coagulation , Blood Platelets/drug effects , Humans , Platelet Function Tests
11.
Thromb Res ; 183: 69-75, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31670229

ABSTRACT

Immune thrombocytopenia (ITP) is a heterogeneous autoimmune disorder characterized by immune-mediated platelet destruction, leading to lower platelet count. Thalidomide is considered as a novel immunomodulatory drug for treating several autoimmune diseases. Whether thalidomide can ameliorate ITP remains unclear. This study aims to evaluate the effect of thalidomide on ITP mouse model. ITP mouse model was established through intraperitoneal injection of rat anti-mouse integrin GPIIb/CD41 immunoglobulin. Thalidomide (10, 20 or 50 mg/kg body weight) was intraperitoneally injected into mice followed by antibody injection. Then, peripheral blood and plasma was isolated for analysis of platelet count and the level of IFN-γ and IL-17 in plasma. Meanwhile, spleen was extracted to measure the expression of CD68, a macrophage marker. In addition, macrophage cell line RAW264.7 was cultured and treated with thalidomide followed by analysis of cell viability, apoptosis as well as cell cycle. Thalidomide prevented antiplatelet antibody-mediated platelet destruction in ITP mouse model. Compared with vehicle (phosphate-buffered saline), thalidomide significantly inhibited the secretion of IFN-γ and IL-17 in ITP mouse and reduced the expression of CD68 in spleen. After thalidomide treatment, the cell viability of RAW264.7 cell was significantly reduced and the cell number in S phase was also significantly decreased. In addition, the expression of cyclin E2 was significantly reduced. In conclusion, thalidomide prevents antiplatelet antibody-mediated platelet destruction in ITP mouse possibly through reducing the number of macrophages, suggesting that it might be a novel approach for treating ITP.


Subject(s)
Immunosuppressive Agents/therapeutic use , Thalidomide/therapeutic use , Thrombocytopenia/drug therapy , Animals , Disease Models, Animal , Immunosuppressive Agents/pharmacology , Mice , Thalidomide/pharmacology , Thrombocytopenia/pathology
12.
Thromb Haemost ; 119(10): 1655-1664, 2019 10.
Article in English | MEDLINE | ID: mdl-31370073

ABSTRACT

All-trans retinoic acid (ATRA) is widely used for induction of complete remission in patients with acute promyelocytic leukemia (APL). ATRA also regulates protein kinase C (PKC) activity. Therapeutic use of ATRA reportedly interferes with hemostatic function in APL patients, including effects on coagulation or other vascular cells, although effects of ATRA on platelets remain unclear. This study aims to investigate the effect of therapeutic-relevant doses of ATRA on platelet function. Human platelets were preincubated with ATRA (0-20 µM) for 1 hour at 37°C, followed by analysis of aggregation, granule secretion, receptor expression by flow cytometry, platelet spreading, or clot retraction. Additionally, ATRA (10 mg/kg) was injected intraperitoneally into mice and tail bleeding time and arterial thrombus formation were evaluated. ATRA inhibited platelet aggregation and adenosine triphosphate release induced by collagen (5 µg/mL) or thrombin (0.05 U/mL) in a dose-dependent manner without affecting P-selectin expression or surface levels of glycoprotein (GP) Ibα, GPVI, or αIIbß3. ATRA-treated platelets demonstrated reduced spreading on immobilized fibrinogen or collagen and reduced thrombin-induced clot retraction together with reduced phosphorylation of Syk and PLCγ2. In addition, ATRA-treated mice displayed significantly impaired hemostasis and arterial thrombus formation in vivo. Further, in platelets stimulated with either collagen-related peptide or thrombin, ATRA selectively inhibited phosphorylation of PKCßI (Ser661) and PKCδ (Thr505), but not PKCα or PKCßII phosphorylation (Thr638/641). In conclusion, ATRA inhibits platelet function and thrombus formation, possibly involving direct or indirect inhibition of PKCßI/δ, indicating that ATRA might be beneficial for the treatment of thrombotic or cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/metabolism , Protein Kinase C beta/metabolism , Protein Kinase C-delta/metabolism , Thrombosis/drug therapy , Tretinoin/pharmacology , Animals , Blood Coagulation/drug effects , Blood Platelets/metabolism , Clot Retraction/drug effects , Collagen/chemistry , Cytoskeleton/metabolism , Hemostasis/drug effects , Humans , Mice , Mice, Inbred C57BL , Phosphorylation , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Function Tests , Signal Transduction/drug effects
13.
Int Immunopharmacol ; 66: 91-98, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30445311

ABSTRACT

Immune thrombocytopenia (ITP) is a heterogeneous autoimmune disease, characterized by accelerated platelet destruction/clearance or decreased platelet production. ADAM17-mediated platelet receptor GPIbα extracellular domain shedding has been shown to be involved in platelet clearance. Whether GPIbα shedding participates in the pathogenesis of ITP remains poorly understood. This study aims to investigate the role of GPIbα shedding in the development of ITP via incubating normal platelets with ITP plasma to mimic ITP in vivo environment. Plasma was isolated from ITP patients or healthy control and incubated with platelets in vitro followed by measuring GPIbα expression by flow cytometry and western blot, ADAM17 expression by western blot, ROS generation and platelet activation by flow cytometry. Compared with control plasma, ITP plasma-treated platelet displayed significantly reduced GPIbα surface expression, increased ADAM17 expression and ROS generation. However, metalloproteinase inhibitor GM6001 blocked the ITP-plasma-induced decrease in GPIbα surface expression, increase in ADAM17 expression and platelet activation. In addition, inhibitors of NADPH oxidase or mitochondria respiration significantly inhibited ROS generation from ITP plasma-treated platelets. Moreover, ROS inhibition or blocking FcγRIIa attenuated the decrease in GPIbα surface expression, platelet activation and ROS generation (for blocking FcγRIIa) in ITP plasma-treated platelets. In conclusion, ITP plasma induces platelet receptor GPIbα extracellular domain shedding, suggesting that it might participate in the pathogenesis of ITP and targeting it might be a novel approach for treating ITP.


Subject(s)
Blood Platelets/immunology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Purpura, Thrombocytopenic, Idiopathic/immunology , ADAM17 Protein/metabolism , Adult , Aged , Cells, Cultured , Down-Regulation , Female , Humans , Male , Metalloproteases/metabolism , Middle Aged , NADPH Oxidases/metabolism , Plasma/immunology , Reactive Oxygen Species/metabolism , Receptors, IgG/metabolism , Young Adult
14.
Int Immunopharmacol ; 55: 63-68, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29223855

ABSTRACT

Immune thrombocytopenia (ITP) is an autoimmune disease, which is characterized by abnormal of T immunity. A disintegrin and metalloproteinase (ADAM) 10, a member of proteinase family, has been demonstrated to regulate T cell proliferation and effector function. Considering the closely association of dysregulation of T cell function with ITP, whether ADAM10 involves in the pathogenesis of ITP remains unclear. In this study, 54 active ITP patients, 18 ITP in remission and 24 age and gender matched healthy control were enrolled. Peripheral blood mononuclear cells (PBMCs) were isolated from patients and control for isolation of RNA and plasma which were used to measure mRNA level of ADAM10 and tissue inhibitor of metalloproteinase 3 (TIMP3) by quantitative real-time PCR and soluble level of FasL and lymphocyte activation gene-3 (LAG-3) in plasma by ELISA. Meanwhile, T cell activation was measured by flow cytometry. Our results showed significantly higher expression of ADAM10 and lower expression of TIMP3 in active ITP patients compared with control, which were all restored into normal level in remission patients. Consistent with the expression profile of ADAM10, increased soluble plasma level of FasL and LAG-3 were observed in active ITP patients and reduced to normal level in patients in remission. Furthermore, increased T cell activation as demonstrated by higher expression of HLA-DR and CD69 were found in active ITP patients. In conclusion, elevated expression of ADAM10 was associated with the pathogenesis and development of ITP and therapeutically targeting it might be a novel approach for the treatment of ITP.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Antigens, CD/metabolism , Membrane Proteins/metabolism , Purpura, Thrombocytopenic, Idiopathic/genetics , T-Lymphocytes/immunology , Tissue Inhibitor of Metalloproteinase-3/metabolism , ADAM10 Protein/genetics , Adolescent , Adult , Aged , Amyloid Precursor Protein Secretases/genetics , Antigens, CD/genetics , Cell Proliferation/genetics , Cells, Cultured , Fas Ligand Protein/blood , Female , Humans , Lymphocyte Activation , Male , Membrane Proteins/genetics , Middle Aged , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/metabolism , Tissue Inhibitor of Metalloproteinase-3/genetics , Up-Regulation , Young Adult , Lymphocyte Activation Gene 3 Protein
15.
Transpl Immunol ; 16(3-4): 166-71, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17138049

ABSTRACT

We have previously shown that a novel immunotherapy using ex vivo activated immune cells is capable of promoting survival and hematopoietic recovery in mice after combined chemotherapy and radiotherapy. In this study, we investigated whether the immunotherapy with ex vivo activated immune cells had the same beneficial effects after syngeneic and semiallogeneic bone marrow transplantation (BMT) in BALB/c mice subjected to a lethal dose of total body irradiation (TBI). Immune cells were cultured in vitro with a combination of cytokines and a calcium ionophore for 2 days and subsequently injected to mice daily for 4 days starting 1 day after BMT. The immunotherapy enhanced survival and multilineage peripheral blood recovery in BMT mice with limited numbers of transplanted bone marrow cells when a low dose of ex vivo activated immune cells were used. However, the beneficial effects were completely lost when a higher dose of the same therapeutic immune cells were tested, and instead the immunotherapy significantly exacerbated complications associated with the lethal radiation and BMT. This detrimental effect appeared to be the result of strong in vivo nonspecific immune responses induced by either activated therapeutic immune cells or interaction between therapeutic immune cells and MHC-mismatched bone marrow cells transplanted or both. Our data suggest that the immunotherapy with appropriately selected dosages may be beneficial to BMT but vigorous in vivo immune responses soon after BMT may exacerbate post-transplant complications.


Subject(s)
Bone Marrow Transplantation/immunology , Graft Rejection/prevention & control , Immunotherapy/methods , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/transplantation , Animals , B-Lymphocytes/immunology , Cell Culture Techniques , Female , Hematopoiesis/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , T-Lymphocytes/immunology , Transplantation, Homologous , Transplantation, Isogeneic
16.
Zhonghua Zhong Liu Za Zhi ; 27(9): 565-6, 2005 Sep.
Article in Zh | MEDLINE | ID: mdl-16438859

ABSTRACT

OBJECTIVE: To investigate the diagnostic value of dual-head (18)F-fluorodeoxyglucose ((18)F-FDG) imaging in metastatic lesion with unknown primary tumour (UPT). METHODS: Seventy patients with UPT underwent dual-head (18)F-FDG imaging after iv (18)F-FDG 1.85 MBq/kg. The primary tumour was diagnosed according to the FDG uptake and T/N value. RESULTS: Of the 70 patients, the primary tumour was identified by positive FDG imaging and finally confirmed pathologically in 58 patients (82.9%), and 12 patients had a negative FDG imaging (17.1%). Forty-two of the 58 positive patients were found to have lung cancer (72.4%). Among the 12 negative patients, their primary tumour was then identified by other diagnostic procedures in 5 patients (41.7%), in 1 patient, the primary site was detected during follow-up, however, the primary tumour was never detected in the rest 6 patients. CONCLUSION: Dual-probe (18)F-FDG imaging is a simple, quick, non-invasive and sensitive technique with an accuracy over 80% in the diagnosis of unknown primary tumour. The lung is found to be the most frequent primary site. Dual-probe (18)F-FDG imaging can be recommended as the first diagnostic choice for UPT.


Subject(s)
Fluorodeoxyglucose F18 , Lung Neoplasms/diagnostic imaging , Lymph Nodes/diagnostic imaging , Neoplasms, Unknown Primary/diagnostic imaging , Positron-Emission Tomography , Female , Humans , Lung Neoplasms/pathology , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Neoplasms, Unknown Primary/pathology
17.
Ai Zheng ; 23(10): 1222-4, 2004 Oct.
Article in Zh | MEDLINE | ID: mdl-15473941

ABSTRACT

BACKGROUND & OBJECTIVE: Nasopharynx applicator used in intracavitary brachytherapy plays an important role in the radiotherapy of nasopharyngeal carcinoma (NPC), its quality affects the efficiency of treatment. This study was to design a new applicator for clinical use. METHODS: An inexpensive, reusable, and flexible latex nasopharynx applicator was designed. An air bag was placed at 15 mm from the foreside of the applicator, clung to the tube. The edge of air bag is tangent to the axis of tube. When the bag was full of air, the tube would hunch reversely,close to nasopharyngeal vault. After introduced into nasopharynx through middle nasal meatus, the applicator could be fixed in suitable position by its rotation, and air bag regulation, and confirmed its position by simulation. RESULTS: A total of 221 patients with NPC were treated with external beam radiation therapy in our hospital, and boosted HDR brachytherapy using this new applicator. The response rate was 92.6% in the primary tumor group (200/216), and 100% in the recurrent tumor group (5/5). Mucosal necrosis in the posterior or anterior wall of nasopharynx occurred in 5 patients, 8 patients experienced nasal congestion and nasal synechia. CONCLUSIONS: This new nasopharynx applicator is easy to operate, painless, and well dosage-distributed. Mucosal necrosis is likely due to higher fractional dose.


Subject(s)
Brachytherapy/instrumentation , Carcinoma, Squamous Cell/radiotherapy , Nasopharyngeal Neoplasms/radiotherapy , Adolescent , Adult , Aged , Brachytherapy/methods , Cobalt Radioisotopes/therapeutic use , Equipment Design , Equipment Safety , Female , Humans , Male , Middle Aged , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL