Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Blood Cells Mol Dis ; 104: 102798, 2024 01.
Article in English | MEDLINE | ID: mdl-37813040

ABSTRACT

Thrombocytopenia is a critical complication after radiation therapy and exposure. Dysfunction of megakaryocyte development and platelet production are key pathophysiological stages in ionizing radiation (IR)-induced thrombocytopenia. Protein kinase C (PKC) plays an important role in regulating megakaryocyte development and platelet production. However, it remains unclear how PKC regulates IR-induced megakaryocyte apoptosis. In this study, we found that pretreatment of PKC pan-inhibitor Go6983 delayed IR-induced megakaryocyte apoptosis, and inhibited IR-induced mitochondrial membrane potential and ROS production in CMK cells. Moreover, suppressing PKC activation inhibited cleaved caspase3 expression and reduced p38 phosphorylation levels, and IR-induced PKC activation might be regulated by p53. In vivo experiments confirmed that Go6983 promoted platelet count recovery after 21 days of 3 Gy total body irradiation. Furthermore, Go6983 reduced megakaryocyte apoptosis, increased the number of megakaryocyte and polyploid formation in bone marrow, and improved the survival rate of 6 Gy total body irradiation. In conclusion, our results provided a potential therapeutic target for IR-induced thrombocytopenia.


Subject(s)
Megakaryocytes , Thrombocytopenia , Humans , Protein Kinase C/metabolism , Protein Kinase C/therapeutic use , X-Rays , Thrombocytopenia/etiology , Thrombopoiesis , Apoptosis , Blood Platelets
2.
Curr Issues Mol Biol ; 45(7): 6024-6039, 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37504296

ABSTRACT

X-rays can induce morphological as well as functional changes in cells. Platelets are anuclear cellular fragments originating from megakaryocytes and are the major regulators in hemostasis and thrombosis. Platelet products are irradiated to avoid medical complications associated with platelet transfusion. So far, gamma, UV, and laser radiation have been used for this purpose. However, scientists are divided about the effects of radiation on platelet quality. The present study was designed to explore the possible effects of X-rays in washed human platelets and understand the molecular mechanism behind them. In the present study, we exposed washed human platelets to 10 or 30 Gy X-rays at 0.25 Gy/min. Flow cytometry, aggregometry, and western blot were performed to investigate the effect of X-rays on platelet degranulation, integrin activation, platelet aggregation, and apoptosis. It was found that X-rays immediately induced granular secretions with no effect on GP IIb/IIIa activation. Not surprisingly, due to granule secretions in irradiated platelets, platelet aggregation was significantly reduced. In contrast to granular secretions and platelet aggregation, X-rays induced mitochondrial transmembrane potential depolarization in a time-dependent manner to induce apoptosis and activated protein kinase C (PKC) signaling. This study revealed and explained the molecular mechanism activated by X-rays in washed human platelets. Here we also introduced Gö 6983, a PKC inhibitor, as an agent that counteracts X-ray-induced changes and maintains the integrity of platelets.

3.
Platelets ; 33(3): 381-389, 2022 Apr 03.
Article in English | MEDLINE | ID: mdl-33979555

ABSTRACT

Glycoprotein (GP) Ibα shedding mediated by ADAM17 (a disintegrin and metalloproteinase 17) plays an important role in negatively regulating platelet function and thrombus formation. However, the mechanism of GPIbα shedding remains elusive. Here, we show that jasplakinolide (an actin-polymerizing peptide)-induced actin polymerization results in GPIbα shedding and impairs platelet function. Thrombin and A23187-induced GPIbα shedding is increased by jasplakinolide; in contrast, GPIbα shedding is reduced by a depolymerization regent (cytochalasin B). We find that actin polymerization activates calpain leading to filamin A hydrolyzation. We further demonstrate that the interaction of filamin A with the cytoplasmic domain of GPIbα plays a critical role in regulating actin polymerization-induced GPIbα shedding. Taken together, these data demonstrate that actin polymerization regulates ADAM17-mediated GPIbα shedding, suggesting a novel strategy to negatively regulate platelet function.


Subject(s)
Actins/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Animals , Healthy Volunteers , Humans , Mice , Polymerization
4.
Proc Natl Acad Sci U S A ; 115(45): E10682-E10691, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30337485

ABSTRACT

Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by low platelet count which can cause fatal hemorrhage. ITP patients with antiplatelet glycoprotein (GP) Ib-IX autoantibodies appear refractory to conventional treatments, and the mechanism remains elusive. Here we show that the platelets undergo apoptosis in ITP patients with anti-GPIbα autoantibodies. Consistent with these findings, the anti-GPIbα monoclonal antibodies AN51 and SZ2 induce platelet apoptosis in vitro. We demonstrate that anti-GPIbα antibody binding activates Akt, which elicits platelet apoptosis through activation of phosphodiesterase (PDE3A) and PDE3A-mediated PKA inhibition. Genetic ablation or chemical inhibition of Akt or blocking of Akt signaling abolishes anti-GPIbα antibody-induced platelet apoptosis. We further demonstrate that the antibody-bound platelets are removed in vivo through an apoptosis-dependent manner. Phosphatidylserine (PS) exposure on apoptotic platelets results in phagocytosis of platelets by macrophages in the liver. Notably, inhibition or genetic ablation of Akt or Akt-regulated apoptotic signaling or blockage of PS exposure protects the platelets from clearance. Therefore, our findings reveal pathogenic mechanisms of ITP with anti-GPIbα autoantibodies and, more importantly, suggest therapeutic strategies for thrombocytopenia caused by autoantibodies or other pathogenic factors.


Subject(s)
Apoptosis , Blood Platelets/cytology , Proto-Oncogene Proteins c-akt/metabolism , Purpura, Thrombocytopenic, Idiopathic/pathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Glycoproteins/immunology , Humans , Liver/metabolism , Macrophages/metabolism , Phagocytosis , Phosphoric Diester Hydrolases/metabolism , Purpura, Thrombocytopenic, Idiopathic/enzymology , Signal Transduction
5.
Proc Natl Acad Sci U S A ; 114(11): 2964-2969, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28242694

ABSTRACT

Previous studies have shown that receptor-interacting protein kinase 3 (RIP3) is involved in many important biological processes, including necroptosis, apoptosis, and inflammation. Here we show that RIP3 plays a critical role in regulating platelet functions and in vivo thrombosis and hemostasis. Tail bleeding times were significantly longer in RIP3-knockout (RIP3-/-) mice compared with their wild-type (WT) littermates. In an in vivo model of arteriole thrombosis, mice lacking RIP3 exhibited prolonged occlusion times. WT mice repopulated with RIP3-/- bone marrow-derived cells had longer occlusion times than RIP3-/- mice repopulated with WT bone marrow-derived cells, suggesting a role for RIP3-deficient platelets in arterial thrombosis. Consistent with these findings, we observed that RIP3 was expressed in both human and mice platelets. Deletion of RIP3 in mouse platelets caused a marked defect in aggregation and attenuated dense granule secretion in response to low doses of thrombin or a thromboxane A2 analog, U46619. Phosphorylation of Akt induced by U46619 or thrombin was diminished in RIP3-/- platelets. Moreover, RIP3 interacted with Gα13 Platelet spreading on fibrinogen and clot retraction were impaired in the absence of RIP3. RIP3 inhibitor dose-dependently inhibited platelet aggregation in vitro and prevented arterial thrombus formation in vivo. These data demonstrate a role for RIP3 in promoting in vivo thrombosis and hemostasis by amplifying platelet activation. RIP3 may represent a novel promising therapeutic target for thrombotic diseases.


Subject(s)
Platelet Activation/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Thrombosis/genetics , Thrombosis/metabolism , Adenosine Diphosphate/metabolism , Animals , Blood Platelets/metabolism , Disease Models, Animal , Gene Expression , Hemostasis/genetics , Humans , Mice , Mice, Knockout , Phosphatidylserines/metabolism , Phosphorylation , Platelet Aggregation/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Thrombin/metabolism , Thromboxane A2/metabolism
6.
Clin Lab ; 64(9): 1413-1419, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30274018

ABSTRACT

BACKGROUND: Patients with Parkinson's disease (PD) are at a lower risk of suffering cardiovascular events, but the underlying factors for this decreased risk remain unclear. Serum triglycerides (TG) and total cholesterol (TC), and their expression relative to high-density lipoprotein cholesterol (TG/HDL-C and TC/HDL-C), are independent predictors of cardiovascular events. This study aimed to determine if PD patients have decreased lipid levels and lipid ratios, which may underlie the decreased risk of coronary heart disease (CHD). METHODS: This retrospective study included 92 PD patients (PD group), 450 control subjects with no CHD (OD group), and 450 CHD patients (CHD group). We analyzed serum lipid levels and lipid ratios in each group. RESULTS: There were significant differences in TC (F = 10.459, p < 0.0001), TG (F = 46.856, p < 0.0001), low density lipoprotein cholesterol (LDL-C) (F = 6.910, p = 0.001), high density lipoprotein cholesterol HDL-C (F = 30.694, p < 0.0001), TC/HDL-C (F = 32.675, p < 0.0001), and TG/HDL-C (F = 45.554, p < 0.0001) between all three groups; TC/LDL-C (F = 2.518, p = 0.081) was not significantly different between groups. Compared to the CHD group, PD patients had lower TC (p < 0.0001), TG (p < 0.0001), LDL-C (p = 0.001), TG/HDL-C (p < 0.0001), and TC/HDL-C (p < 0.0001); TC/LDL-C (p = 0.563) and HDL-C (p = 0.196) were not significantly different. TC and LDL-C levels were positively correlated within individual groups (all p < 0.0001). In addition, TG and HDL-C were negatively correlated in the OD and CHD groups (p < 0.0001); no significant negative association was observed in the PD group (p = 0.077). TG/HDL and LDL-C levels were inversely correlated in the CHD group (p < 0.0001) and weakly positively correlated in the PD (p = 0.159) and OD (p = 0.199) groups. CONCLUSIONS: TC/HDL and TG/HDL ratios were significantly lower in PD patients compared to CHD patients, suggesting there is a strong correlation between lipid ratios and incidence of CHD in PD patients.


Subject(s)
Coronary Disease/blood , Coronary Disease/epidemiology , Lipids/blood , Parkinson Disease/blood , Parkinson Disease/epidemiology , Aged , Biomarkers/blood , China/epidemiology , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Coronary Disease/diagnosis , Down-Regulation , Female , Humans , Incidence , Male , Middle Aged , Parkinson Disease/diagnosis , Prevalence , Prognosis , Retrospective Studies , Risk Factors , Triglycerides/blood
7.
Alcohol Clin Exp Res ; 41(2): 291-298, 2017 02.
Article in English | MEDLINE | ID: mdl-28081301

ABSTRACT

BACKGROUND: Alcohol abuse incurs severe medical conditions, such as thrombocytopenia and hemorrhage, but the pathogenesis is not totally understood. Alcohol has been reported to induce apoptosis in eukaryotic cells, such as hepatocyte, nerve cell, corneal fibroblasts. However, it is still unclear whether alcohol induces platelet apoptosis. METHODS: Washed human platelets were pretreated with ethanol (EtOH), and apoptotic events and platelet function were detected. In in vivo experiments, C57BL/6J mice were given EtOH by gavage. Platelet counts, tail bleeding time, and the stomach were examined. RESULTS: EtOH dose dependently induces depolarization of mitochondrial inner transmembrane potential, up-regulation of Bax, down-regulation of Bcl-2, and caspase-3 activation. EtOH does not induce surface expression of P-selectin or PAC-1 binding, whereas significantly reduces collagen-, thrombin-, and ADP-induced platelet aggregation. Moreover, EtOH induces c-Jun NH2-terminal kinase activation. In an in vivo mouse model of the acute alcoholism, EtOH significantly reduces the number of circulating platelets, prolongs the tail bleeding time, and causes gastric mucosa hemorrhage. CONCLUSIONS: These data demonstrate that EtOH induces mitochondria-mediated intrinsic platelet apoptosis, results in the reduction of the number of circulating platelets, and impairs in vivo hemostasis. These findings reveal the possible pathogenesis of hemorrhagic symptoms in patients experiencing acute alcohol intoxication.


Subject(s)
Apoptosis/drug effects , Blood Platelets/drug effects , Central Nervous System Depressants/toxicity , Ethanol/toxicity , Animals , Bleeding Time , Caspase 3/biosynthesis , Dose-Response Relationship, Drug , Gastrointestinal Hemorrhage/chemically induced , Humans , In Vitro Techniques , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Platelet Count , Platelet Function Tests , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
8.
Environ Toxicol ; 31(2): 163-75, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25059219

ABSTRACT

Zebrafish (Danio rerio) has been used historically for evaluating the toxicity of environmental and aqueous toxicants, and there is an emerging literature reporting toxic effects of manufactured nanoparticles (NPs) in zebrafish embryos. Few researches, however, are focused on the neurotoxicity on adult zebrafish after subchronic exposure to TiO2 NPs. This study was designed to evaluate the morphological changes, alterations of neurochemical contents, and expressions of memory behavior-related genes in zebrafish brains caused by exposures to 5, 10, 20, and 40 µg/L TiO2 NPs for 45 consecutive days. Our data indicated that spatial recognition memory and levels of norepinephrine, dopamine, and 5-hydroxytryptamine were significantly decreased and NO levels were markedly elevated, and over proliferation of glial cells, neuron apoptosis, and TiO2 NP aggregation were observed after low dose exposures of TiO2 NPs. Furthermore, the low dose exposures of TiO2 NPs significantly activated expressions of C-fos, C-jun, and BDNF genes, and suppressed expressions of p38, NGF, CREB, NR1, NR2ab, and GluR2 genes. These findings imply that low dose exposures of TiO2 NPs may result in the brain damages in zebrafish, provide a developmental basis for evaluating the neurotoxicity of subchronic exposure, and raise the caution of aquatic application of TiO2 NPs.


Subject(s)
Nanoparticles/toxicity , Neurotoxicity Syndromes/pathology , Titanium/toxicity , Zebrafish , 5-Hydroxytryptophan/metabolism , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Body Weight/drug effects , Brain/pathology , Brain/ultrastructure , Cell Proliferation/drug effects , Dopamine/metabolism , Gene Expression/drug effects , Memory/drug effects , Neuroglia/drug effects , Neurotoxicity Syndromes/psychology , Nitric Oxide/metabolism , Norepinephrine/metabolism , Recognition, Psychology/drug effects
9.
Environ Toxicol ; 29(1): 64-73, 2014 Jan.
Article in English | MEDLINE | ID: mdl-21928445

ABSTRACT

In an effort to investigate the effects of exposure to lanthanoids (Ln) on the immune response and liver function, mice were orally exposed to LaCl3 , CeCl3 , and NdCl3 at 2, 10, and 20 mg/kg doses for 30 days, respectively; lymphocyte counts, serum IgM level, hematological indices, biochemical parameters of liver functions, and histopathological changes in Ln(3+) -treated mice were assessed. Indeed, 20 mg/kg Ln(3+) significantly inhibited mice growth and reduced the counts of white blood cells, platelets, and reticulocyte in mice blood. Specifically, in these Ln(3+) -treated mice, CD3+, CD4+, CD8+, CD19+ and NK cells, and CD4+/CD8+ ratio as well as serum IgM level were decreased. Furthermore, liver function was disrupted, as evidenced by the increased alanine aminotransferase, total bilirubin, total bile acid and triglycerides, and the decreased glucose and ratio of albumin to globulin. The cytoarchitecture damage and fatty degeneration in liver caused by Ln(3+) at 20 mg/kg dose were also observed. Our findings showed that exposure to Ln affected the cell and humoral immunity and disturbed liver function in mice. In addition, Ce(3+) was found to exhibit higher toxicity than La(3+) and Nd(3+).


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Immune System/drug effects , Lanthanoid Series Elements/toxicity , Liver/drug effects , Animals , Antigens, CD/immunology , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Female , Immune System/immunology , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunoglobulin M/blood , Immunoglobulin M/immunology , Liver/immunology , Liver/pathology , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Male , Mice , Random Allocation
10.
Environ Toxicol ; 29(7): 837-46, 2014 May.
Article in English | MEDLINE | ID: mdl-23139204

ABSTRACT

Numerous studies have demonstrated lanthanide (Ln) accumulation in the liver, and the corresponding damage; however, very little work has been done to evaluate the relationship between Ln-induced liver injury and its gene expression profile in mice. In this study, liver injury and gene-expressed profiles in male mice induced by oral administration of CeCl3 (2 mg/kg) via gavage for 90 consecutive days were investigated. The results showed that cerium accumulation, liver inflammation, and hepatocyte necrosis were observed. CeCl3 exposure significantly decreased the counts of white blood cells, lymphocyte, and platelet, the reticulocyte count (Ret) and neutrophilic granulocyte percentages as well as A/G ratio, whereas markedly increased the activities of alkaline phosphatase, lactate dehydrogenase, and cholinesterase, and the concentrations of triglycerides and total cholesterol. Furthermore, microarray results of liver showed that the differential expression of 675 known function genes involved in immune/inflammation response, apoptosis, metabolic process, cell cycle, cell proliferation, cytoskeleton, oxidative stress, signal transduction, transcription, translation, and transportation in CeCl3 exposed livers, respectively. Specifically, the significant downregulation of Nt5e led to inflammation, overexpressed Cyp4a12a and great suppression of Cdkn1a resulted in hepatocyte apoptosis, marked elevation of Cel, and Cyp7b1 expression caused the metabolic disorders in mouse liver after long-term CeCl3 exposure. Therefore, these genes may be in great relation to liver damages induced by exposure to CeCl3 .


Subject(s)
Cerium/toxicity , Environmental Pollutants/toxicity , Liver/drug effects , Animals , Apoptosis/genetics , Biomarkers/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Liver/metabolism , Liver/pathology , Liver Function Tests , Male , Mice, Inbred ICR , Oxidative Stress/drug effects , Transcriptome
11.
Part Fibre Toxicol ; 10: 4, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23406204

ABSTRACT

BACKGROUND: Numerous studies have demonstrated that titanium dioxide nanoparticles (TiO2 NPs) induced nephrotoxicity in animals. However, the nephrotoxic multiple molecular mechanisms are not clearly understood. METHODS: Mice were exposed to 2.5, 5 and 10 mg/kg TiO2 NPs by intragastric administration for 90 consecutive days, and their growth, element distribution, and oxidative stress in kidney as well as kidney gene expression profile were investigated using whole-genome microarray analysis technique. RESULTS: Our findings suggest that TiO2 NPs resulted in significant reduction of renal glomerulus number, apoptosis, infiltration of inflammatory cells, tissue necrosis or disorganization of renal tubules, coupled with decreased body weight, increased kidney indices, unbalance of element distribution, production of reactive oxygen species and peroxidation of lipid, protein and DNA in mouse kidney tissue. Furthermore, microarray analysis showed significant alterations in the expression of 1, 246 genes in the 10 mg/kg TiO2 NPs-exposed kidney. Of the genes altered, 1006 genes were associated with immune/inflammatory responses, apoptosis, biological processes, oxidative stress, ion transport, metabolic processes, the cell cycle, signal transduction, cell component, transcription, translation and cell differentiation, respectively. Specifically, the vital up-regulation of Bcl6, Cfi and Cfd caused immune/ inflammatory responses, the significant alterations of Axud1, Cyp4a12a, Cyp4a12b, Cyp4a14, and Cyp2d9 expression resulted in severe oxidative stress, and great suppression of Birc5, Crap2, and Tfrc expression led to renal cell apoptosis. CONCLUSIONS: Axud1, Bcl6, Cf1, Cfd, Cyp4a12a, Cyp4a12b, Cyp2d9, Birc5, Crap2, and Tfrc may be potential biomarkers of kidney toxicity caused by TiO2 NPs exposure.


Subject(s)
Gene Expression/drug effects , Kidney Diseases/chemically induced , Kidney/drug effects , Nanoparticles/toxicity , Titanium/toxicity , Administration, Oral , Animals , Apoptosis/drug effects , Body Weight/drug effects , Dose-Response Relationship, Drug , Gene Expression Profiling , Kidney/metabolism , Kidney/ultrastructure , Kidney Diseases/genetics , Kidney Diseases/pathology , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred Strains , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Oxidative Stress/drug effects , Particle Size , Surface Properties , Titanium/administration & dosage , Titanium/chemistry
12.
Environ Toxicol ; 28(6): 349-58, 2013 Jun.
Article in English | MEDLINE | ID: mdl-21656643

ABSTRACT

To investigate the molecular mechanism of inflammatory response in the mouse liver caused by exposure to CeCl3, we measured the liver indices, and cerium content, evaluated the liver histopathological section, detected serum biochemical parameters of liver function, and the immunoglobulin M (IgM) content, analyzed the liver mRNA and protein expression levels of Toll-like receptor 2, 4 (TLR2, TLR4), and inflammatory cytokines in liver using real-time quantitative reverse transcriptase polymerase chain reaction and enzyme-linked immunosorbent assay. The results showed that exposure to CeCl3 decreased body weight and caused cerium accumulation in the mouse liver and histopathological changes of liver (such as inflammatory cell infiltration). Furthermore, biochemical assays suggested that CeCl3 could promote the activities of alanine aminotransferase, alkaline phosphatase, aspartate aminotransferase, lactate dehydrogenase, pseudocholinesterase, and leucine aminopeptidase, decrease serum IgM, upregulate the levels of TLR2, TLR4, nuclear factor-κB (NF-κB), NF-κBp52, NF-κBp65, NF-κB-inducing kinase (NIK), IκB kinase α (IKK-α), IκB kinase ß (IKK-ß), and tumor necrosis factor-α (TNF-α) expression, and suppress NF-κB-inhibiting factor (IκB) and interleukin-2 (IL-2) expression in liver. Taken together, the inflammation of mice liver caused by exposure to CeCl3 might be closely associated with the alteration of inflammatory cytokine expressions in the mouse liver, the signal-transducing events happening in CeCl3-induced macrophages of liver sequentially might occur via activation of TLRs→TNF-α→NIK→IκB kinase (including IKK1, IKK2)→NF-κB (including NF-κBP52, NF-κBP65)→ inflammation.


Subject(s)
Cerium/toxicity , Liver/metabolism , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Animals , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Immunoglobulin M/blood , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Liver/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred ICR , Signal Transduction/drug effects , Toll-Like Receptors/metabolism
13.
Environ Toxicol ; 27(12): 707-18, 2012 Dec.
Article in English | MEDLINE | ID: mdl-21384496

ABSTRACT

Experimental studies have demonstrated that lanthanides could impair cognitive functions of children and animals, but very little is known about the hippocampal apoptosis and its molecular mechanism. The study investigated the signal pathway of hippocampal apoptosis induced by intragastric administration of CeCl(3) for 60 consecutive days. It showed that cerium had been significantly accumulated in the mouse hippocampus, and CeCl(3) caused hippocampal apoptosis and impairment of spatial recognition memory of mice. CeCl(3) effectively activated caspase-3 and -9, inhibited Bcl-2, and increased the levels of Bax and cytochrome c, promoted accumulation of reactive oxygen species in the mouse hippocampus. It implied that CeCl(3)-induced apoptosis in the mouse hippocampus could be triggered via mitochondrion-mediated pathway. Our findings suggest the need for great caution to handle the lanthanides for workers and consumers.


Subject(s)
Apoptosis/drug effects , Cerium/toxicity , Hippocampus/drug effects , Memory/drug effects , Signal Transduction/drug effects , Animals , Antioxidants/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cytochromes c/metabolism , Hippocampus/pathology , Lipid Peroxidation , Male , Mice , Mice, Inbred ICR , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/metabolism
14.
Arch Environ Contam Toxicol ; 62(1): 154-64, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21503700

ABSTRACT

Cerium has been demonstrated to damage liver of mice, but very little is known about the molecular mechanisms underlying the mouse liver apoptosis. In order to understand the liver injury induced by intragastric administration of cerium chloride (CeCl3) for 60 consecutive days, the hepatocyte ultrasrtucture, various oxidative stress parameters, and the stress-related gene expression levels were investigated for the mouse liver. The results demonstrated that CeCl3 had an obvious accumulation in the mouse liver, leading to a classical laddering cleavage of DNA and hepatocyte apoptosis. CeCl3 significantly promoted the accumulation of reactive oxygen species and inhibited the stress-related gene expression of superoxide dismutase, catalase, glutathione peroxidase, metallothionein, heat-shock protein 70, glutathione-S-transferase, P53, and transferring, and it effectively activated the cytochrome p450 1A. It implied that CeCl3 resulted in apoptosis and alteration of expression levels of the genes related with metal detoxification/metabolism regulation and radical scavenging action in mice.


Subject(s)
Cerium/toxicity , Chemical and Drug Induced Liver Injury , Environmental Pollutants/toxicity , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Biomarkers/metabolism , Cerium/pharmacokinetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Environmental Pollutants/pharmacokinetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Inactivation, Metabolic , Lipid Peroxidation , Male , Mice , Mice, Inbred ICR , Oxidative Stress , Random Allocation , Reactive Oxygen Species/metabolism
16.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 28(3): 899-903, 2020 Jun.
Article in Zh | MEDLINE | ID: mdl-32552955

ABSTRACT

OBJECTIVE: To investigate the effect of protein kinase A (PKA) activation on aggregation funetion of platelets in vitro. METHODS: The peripheral blood of healthy adults were collected, and the washed platelets were gained from collected peripheral blood. The washed platelets were treated with PKA activator Forskolin, then the platelet aggregation was induced by using Ristocetin, Thrombin, Collagen and ADP respectively, the platelet aggregation level was detected by the platelet aggregator. RESULTS: Compared with the controls, 5 µmol/L forskolin significantly inhibited ADP and collagen-induced platelet aggregation (P<0.001), and showed mild inhibiting effect on Thrombin-induced platelet aggregation (P<0.05). 2.5-10 µmol/L forskolin significantly inhibited ADP and Collagen -induced platelet aggregation (P<0.001); but not showed significantly inhibitory effects on Ristocetin-induced platelet aggregation (P>0.05). CONCLUSION: PKA activation inhibits agonists-induced platelet aggregation.


Subject(s)
Blood Platelets , Platelet Aggregation , Cyclic AMP-Dependent Protein Kinases , Humans , Platelet Aggregation Inhibitors , Ristocetin , Thrombin
17.
J Clin Invest ; 127(12): 4338-4351, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29083324

ABSTRACT

Apoptosis delimits platelet life span in the circulation and leads to storage lesion, which severely limits the shelf life of stored platelets. Moreover, accumulating evidence indicates that platelet apoptosis provoked by various pathological stimuli results in thrombocytopenia in many common diseases. However, little is known about how platelet apoptosis is initiated or regulated. Here, we show that PKA activity is markedly reduced in platelets aged in vitro, stored platelets, and platelets from patients with immune thrombocytopenia (ITP), diabetes, and bacterial infections. Inhibition or genetic ablation of PKA provoked intrinsic programmed platelet apoptosis in vitro and rapid platelet clearance in vivo. PKA inhibition resulted in dephosphorylation of the proapoptotic protein BAD at Ser155, resulting in sequestration of prosurvival protein BCL-XL in mitochondria and subsequent apoptosis. Notably, PKA activation protected platelets from apoptosis induced by storage or pathological stimuli and elevated peripheral platelet levels in normal mice and in a murine model of ITP. Therefore, these findings identify PKA as a homeostatic regulator of platelet apoptosis that determines platelet life span and survival. Furthermore, these results suggest that regulation of PKA activity represents a promising strategy for extending platelet shelf life and has profound implications for the treatment of platelet number-related diseases and disorders.


Subject(s)
Apoptosis , Blood Platelets/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Animals , Bacterial Infections/enzymology , Bacterial Infections/genetics , Bacterial Infections/pathology , Blood Platelets/pathology , Cyclic AMP-Dependent Protein Kinases/genetics , Diabetes Mellitus/enzymology , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Disease Models, Animal , Enzyme Activation/genetics , Female , Humans , Male , Mice , Mice, Knockout , Purpura, Thrombocytopenic, Idiopathic/enzymology , Purpura, Thrombocytopenic, Idiopathic/genetics , Purpura, Thrombocytopenic, Idiopathic/pathology , bcl-X Protein/genetics , bcl-X Protein/metabolism
18.
Environ Toxicol Pharmacol ; 42: 69-75, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26773364

ABSTRACT

Statins are widely used in the prevention of atherosclerosis and treatment of coronary artery disease because of pleiotropic effects on thrombosis. Thrombocytopenia and hemorrhage occurred in some statin-treated patients, but the reason remains unclear. In the current study, we show that lovastatin dose-dependently induces depolarization of mitochondrial inner transmembrane potential, leading to up-regulation of Bak, down-regulation of Bcl-XL, and activation of caspase-3/8/9. Lovastatin treatment did not increase the surface expression of P-selectin or PAC-1 binding but led to strongly reduced collagen- and thrombin-induced platelet aggregation. The integrin αIIbß3 antagonist, RGDS, inhibited lovastatin-induced apoptosis in both human platelets and Chinese hamster ovary (CHO) cells stably expressing integrin αIIbß3. The number of circulating platelets in mice was significantly reduced after intraperitoneal injections with lovastatin. Taken together, these data indicate that lovastatin induced caspase-dependent platelet apoptosis. Lovastatin does not incur platelet activation, whereas impairs platelet function and reduces circulating platelets in vivo, suggesting the possible pathogenesis of thrombocytopenia and hemorrhage in patients treated with statins.


Subject(s)
Anticholesteremic Agents/pharmacology , Lovastatin/pharmacology , Animals , Apoptosis , Blood Platelets , CHO Cells , Caspase 3/metabolism , Cricetinae , Cricetulus , Down-Regulation , Membrane Potential, Mitochondrial/drug effects , P-Selectin/metabolism , Platelet Aggregation , Up-Regulation
19.
J Hazard Mater ; 278: 180-8, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24968254

ABSTRACT

Titanium dioxide nanoparticles (TiO2 NPs) have been widely used in various areas, and its potential toxicity has gained wide attention. However, the molecular mechanisms of multiple genes working together in the TiO2 NP-induced splenic injury are not well understood. In the present study, 2.5, 5, or 10mg/kg body weight TiO2 NPs were administered to the mice by intragastric administration for 90 consecutive days, their immune capacity in the spleen as well as the gene-expressed characteristics in the mouse damaged spleen were investigated using microarray assay. The findings showed that with increased dose, TiO2 NP exposure resulted in the increases of spleen indices, immune dysfunction, and severe macrophage infiltration as well as apoptosis in the spleen. Importantly, microarray data showed significant alterations in the expressions of 1041 genes involved in immune/inflammatory responses, apoptosis, oxidative stress, stress responses, metabolic processes, ion transport, signal transduction, cell proliferation/division, cytoskeleton and translation in the 10 mg/kg TiO2 NP-exposed spleen. Specifically, Cyp2e1, Sod3, Mt1, Mt2, Atf4, Chac1, H2-k1, Cxcl13, Ccl24, Cd14, Lbp, Cd80, Cd86, Cd28, Il7r, Il12a, Cfd, and Fcnb may be potential biomarkers of spleen toxicity following exposure to TiO2 NPs.


Subject(s)
Nanoparticles/toxicity , Spleen/drug effects , Titanium/toxicity , Animals , Blood Cell Count , Female , Gene Expression Profiling , Mice, Inbred ICR , Microarray Analysis , Microscopy, Electron, Transmission , Organ Size/drug effects , RNA/genetics , RNA/metabolism , Spleen/metabolism , Spleen/pathology , Spleen/ultrastructure , Titanium/metabolism , Titanium/pharmacokinetics
20.
J Biomed Mater Res A ; 102(2): 470-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23533084

ABSTRACT

Titanium dioxide nanoparticles (TiO2 NPs) are widely used in toothpastes, sunscreens, and products for cosmetic purpose that the human use daily. Although the neurotoxicity induced by TiO2 NPs has been demonstrated, very little is known about the molecular mechanisms underlying the brain cognition and behavioral injury. In this study, mice were exposed to 2.5, 5, and 10 mg/kg body weight (BW) TiO2 NPs by nasal administration for 90 consecutive days, respectively, and their brains' injuries and brain gene-expressed profile were investigated. Our findings showed that TiO2 NPs could be translocated and accumulated in brain, led to oxidative stress, overproliferation of all glial cells, tissue necrosis as well as hippocampal cell apoptosis. Furthermore, microarray data showed significant alterations in the expression of 249 known function genes, including 113 genes upregulation and 136 genes downregulation following exposure to 10 mg/kg BW TiO2 NPs, which were associated with oxidative stress, immune response, apoptosis, memory and learning, brain development, signal transduction, metabolic process, DNA repair, response to stimulus, and cellular process. Especially, significant increases in Col1a1, serine/threonine-protein kinase 1, Ctnnb1, cysteine-serine-rich nuclear protein-1, Ddit4, Cyp2e1, and Krev interaction trapped protein 1 (Krit1) expressions and great decreases in DA receptor D2, Neu1, Fc receptor-like molecules, and Dhcr7 expressions following long-term exposure to TiO2 NPs resulted in neurogenic disease states in mice. Therefore, these genes may be potential biomarkers of brain toxicity caused by TiO2 NPs exposure, and the application of TiO2 NPs should be carried out cautiously.


Subject(s)
Brain Injuries/metabolism , Gene Expression Regulation/drug effects , Nanoparticles/adverse effects , Nerve Tissue Proteins/biosynthesis , Neurotoxicity Syndromes/metabolism , Sunscreening Agents/adverse effects , Titanium/adverse effects , Animals , Behavior, Animal/drug effects , Brain/metabolism , Brain/pathology , Brain Injuries/chemically induced , Brain Injuries/pathology , Cell Proliferation/drug effects , Female , Gene Expression Profiling , Humans , Mice , Mice, Inbred ICR , Nanoparticles/administration & dosage , Neuroglia/metabolism , Neuroglia/pathology , Neurotoxicity Syndromes/pathology , Oxidative Stress/drug effects , Sunscreening Agents/pharmacology , Titanium/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL