Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 704: 149661, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38417343

ABSTRACT

To date only four recombinant growth factors, including Filgrastim (rhG-CSF), have been approved by FDA as radiomitigators to ameliorate hematopoietic acute radiation syndrome (H-ARS). These approved agents are not stable under room-temperature, needing to be stored at 2-8 °C, and would not be feasible in a mass casualty scenario where rapid and cost-effective intervention is crucial. Delta-tocotrienol (δ-T3H), the most potent G-CSF-inducing agent among vitamin E isoforms, exhibited efficiency and selectivity on G-CSF production in comparison with TLR and STING agonists in mice. Five-dose δ-T3H was utilized as the optimal therapeutic regimen due to long-term G-CSF production and the best peripheral blood (PB) recovery of irradiated mice. Comparable with rhG-CSF, sequential administration of δ-T3H post-irradiation improved hematologic recovery and accelerated the regeneration of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) in the bone marrow (BM) and spleen of 6.5Gy irradiated mice; and consistently enhanced repopulation of BM-HSCs. In 4.0Gy irradiated nonhuman primates, δ-T3H exhibited comparable efficacy as rhG-CSF to promote PB recovery and colony-formation of BM-HPCs. Altogether, we demonstrated that sequential administration of delta-tocotrienol ameliorates radiation-induced myelosuppression in mice and non-human primates through inducing G-CSF production, indicated δ-T3H as a promising radiomitigator for the management of H-ARS, particularly in a mass casualty scenario.


Subject(s)
Bone Marrow , Hematopoietic Stem Cells , Vitamin E , Animals , Mice , Bone Marrow/pathology , Bone Marrow/radiation effects , Granulocyte Colony-Stimulating Factor/drug effects , Granulocyte Colony-Stimulating Factor/metabolism , Primates , Recombinant Proteins/pharmacology , Vitamin E/analogs & derivatives , Vitamin E/therapeutic use
2.
Environ Toxicol ; 38(2): 403-414, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36282901

ABSTRACT

This study aimed to explore whether vitamin B complex (folic acid, B6 , and B12 ) could avert DNA methylation changes associated with inflammation induced by acute PM2.5 exposure. Sprague-Dawley rats were administered by gavage with different concentrations of vitamin B complex once a day for 28 days, and then by intratracheal instillation with saline or PM2.5 once every 2 days for three times. Vitamin B continued to be taken during the PM2.5 exposure. Rats were sacrificed 24 h after the last exposure. The results showed that vitamin B complex could block the pathological changes and injury in lungs induced by PM2.5 . Meanwhile, vitamin B complex could prevent the abnormal DNA methylation of IL-4 and IFN-γ to antagonize the imbalance of IL-4/IFN-γ associated with inflammation. It was further found that vitamin B complex could regulate DNA methyltransferases (DNMTs) and increase the S-adenosylmethionine (SAM)/S-Adenosyl-L-homocysteine (SAH) ratio to reverse the hypomethylation of genomic DNA and the abnormal DNA methylation of IL-4 and IFN-γ. In conclusion, vitamin B complex has a protective effect on acute lung injury by attenuating abnormal DNA methylation induced by PM2.5 in rats. This study may provide a new insight into the physiological function of vitamin B to prevent the health effects induced by PM2.5 .


Subject(s)
Acute Lung Injury , DNA Methylation , Lung Injury , Particulate Matter , Vitamin B Complex , Animals , Rats , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Dust , Folic Acid , Inflammation/pathology , Interleukin-4/genetics , Lung/pathology , Lung Injury/chemically induced , Lung Injury/genetics , Particulate Matter/toxicity , Rats, Sprague-Dawley , S-Adenosylmethionine/toxicity , Vitamin B Complex/pharmacology
3.
Mol Cell Biochem ; 391(1-2): 85-94, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24615392

ABSTRACT

Thrombopoietin (TPO) can regulate hematopoiesis and megakaryopoiesis via activation of its receptor, c-Mpl, and multiple downstream signal transduction pathways. Using the cytoplasmic domain of Mpl as bait, we performed yeast two-hybrid screening, and found that the protein Atp5d might associate with Mpl. Atp5d is known as the δ subunit of mitochondrial ATP synthase, but little is known about the function of dissociative Atp5d. The interaction between Mpl and Atp5d was confirmed by the yeast two-hybrid system, mammalian two-hybrid assay, pull-down experiment, and co-immunoprecipitation study in vivo and in vitro. An additional immunofluorescence assay showed that the two proteins can colocalize along the plasma membrane in the cytoplasm. Using the yeast two-hybrid system, we tested a series of cytoplasmic truncated mutations for their ability to bind Atp5d and found an association between Atp5d and the Aa98-113 domain of Mpl. The dissociation of Atp5d from Mpl after TPO stimulation suggests that Atp5d may be a new component of TPO signaling.


Subject(s)
Proton-Translocating ATPases/metabolism , Receptors, Thrombopoietin/metabolism , Animals , Cell Line , Humans , Intracellular Space/metabolism , Mice , Mitochondrial Proton-Translocating ATPases , Protein Binding/drug effects , Protein Interaction Domains and Motifs , Protein Interaction Maps/drug effects , Protein Structure, Tertiary , Receptors, Thrombopoietin/chemistry , Reproducibility of Results , Thrombopoietin/pharmacology , Two-Hybrid System Techniques
4.
PLoS One ; 7(12): e51536, 2012.
Article in English | MEDLINE | ID: mdl-23251567

ABSTRACT

BACKGROUND: Total steroidal saponins extracted from the rhizome of Paris polyphylla Sm. var. yunnanensis (TSSPs) have been widely used in China for the treatment of abnormal uterine bleeding. We previously studied the main active constituents of TSSPs and their structure-activity relationships with respect to rat myometrial contractions. Tg (pennogenin tetraglycoside) was identified as one of the active ingredients in TSSPs able to induce rat myometrial contractions. However, the mechanisms underlying the pharmacological actions on uterine activity have not been described clearly. METHODS: Here Tg was screened for effects on contractile activity in isolated uterine strips from estrogen-primed rats and on MLC20 phosphorylation and related signaling pathways in cultured rat myometrial cells as determined by Western blot. Intracellular calcium ([Ca(2+)](i)) was monitored under a confocal microscope using Fluo-4 AM-loaded myometrial cells. RESULTS: Tg dose-dependently stimulated rat myometrial contractions as well as MLC20 phosphorylation in vitro, which could be completely suppressed by an inhibitor of myosin light chain kinase (MLCK). Use of Ca(2+) channel blockers and kinase inhibitors demonstrated that Tg-induced myometrial contractions are mediated by activation of the phospholipase C (PLC)-inositol triphosphate (IP3) signaling pathway, resulting in increased MLC20 phosphorylation. Furthermore, Y27632, a specific inhibitor of Rho kinase (ROK), notably suppressed Tg-stimulated myometrial contractions and decreased MLC20 phosphorylation. CONCLUSIONS: These data provide evidence that rat myometrial contractility induced by Tg results from enhanced MLC20 phosphorylation, while both PLC-IP3 and RhoA/ROK signaling pathways mediate the process. These mechanisms may be responsible for the therapeutic effects of TSSPs on abnormal uterine bleeding.


Subject(s)
Inositol 1,4,5-Trisphosphate/metabolism , Myosin Light Chains/metabolism , Spirostans/pharmacology , Type C Phospholipases/metabolism , Uterine Contraction/drug effects , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Calcium/metabolism , Calmodulin/metabolism , Dose-Response Relationship, Drug , Female , In Vitro Techniques , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spirostans/chemistry , Time Factors
5.
Thromb Res ; 129(5): e209-16, 2012 May.
Article in English | MEDLINE | ID: mdl-22401797

ABSTRACT

BACKGROUND: Total steroidal saponins extracted from the rhizome of Paris polyphylla Sm. var. yunnanensis (TSSPs) have been demonstrated to promote hemostasis in vivo and induce platelet aggregation in vitro. Pennogenin tetraglycoside (Tg) has been identified as one of the active ingredients in TSSPs and can induce rat platelet aggregation. OBJECTIVE: To investigate the functional role of Tg in platelets and the signaling pathway mechanisms which mediate Tg-induced platelet aggregation. METHODS AND RESULTS: Using scanning electron microscopy, the turbidimetric method and flow cytometry, we demonstrated that Tg induces shape change and concentration-dependently induces aggregation, dense granule secretion and a-granule secretion in rat platelets. The activation characteristics were comprehensively confirmed using transmission electron microscopy. Apyrase and antagonists of the platelet adenosine diphosphate (ADP) receptors, P2Y1 and P2Y12, completely inhibited Tg-induced platelet aggregation, which was not sensitive to indomethacin or SQ29548 inhibition. Furthermore, ADP receptor antagonists inhibited Tg-induced a-granule secretion, and blockade of the P2Y1 receptor prevented Tg-induced platelet shape changes. Tg-induced dense granule secretion was not affected by ADP receptor antagonists or various various pharmacological inhibitors of the intracellular effectors involved in dense granule secretion signaling pathways. CONCLUSION: We identified that Tg directly induces platelet activation and demonstrated that Tg-induced platelet activation depends on dense granule secretion of ADP, which in turn activates the P2Y1 and P2Y12 receptor signaling pathways.


Subject(s)
Blood Platelets/drug effects , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Receptors, Purinergic P1/blood , Spirostans/pharmacology , Animals , Blood Platelets/metabolism , Blood Platelets/physiology , Cardiac Glycosides/blood , Male , Platelet Activation/physiology , Platelet Aggregation/physiology , Rats , Rats, Wistar , Signal Transduction/drug effects , Spirostans/blood
6.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 19(4): 991-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21867631

ABSTRACT

This study was purposed to evaluate the effects of recombinant human granulocyte colony-stimulating factor (rhG-CSF) on hematopoietic reconstruction and survival in beagles exposed to mixed fission neutron and γ-ray. 13 beagles were unilaterally exposed to single dose of 2.3 Gy 90% neutrons. The experiments were divided into 3 groups: irradiation control group (no any treatment, n = 4), supportive care group (n = 5) and rhG-CSF plus supportive care group (n = 4, abbreviated as rhG-CSF group) in which the beagles were subcutaneously injected with 200 µg/kg of rhG-CSF early at half an hour and 24 hours post-irradiation respectively. The results showed that 2.3 Gy 90% neutron irradiation induced a severe acute radiation sickness of bone marrow type. The administration of rhG-CSF increased the survival rate from 60% in supportive care group to 100%. Twice injection of rhG-CSF in the first 24 hours reduced duration of neutropenia, enhanced neutrophil nadir and promoted neutrophil recovery when compared with control cohort administered clinical support. The number of colony-forming cells (CFU-GM, CFU-E, and BFU-E) in peripheral blood of rhG-CSF treated canines increased 2-to 5-fold relative to those of the supportive care group on day 3. All canines treated with rhG-CSF achieved hematopoietic reconstruction as evidenced by the pathological section of sternum while severe shortage of hemopoietic cells remained in the cohorts given supportive care alone. It is concluded that the combination of supportive care and high-dose rhG-CSF can accelerate hematopoietic recovery and enhance survival of dogs exposed to 2.3 Gy mixed neutron and gamma ray.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic System/drug effects , Hematopoietic System/radiation effects , Animals , Dogs , Gamma Rays/adverse effects , Granulocyte Colony-Stimulating Factor/administration & dosage , Neutron Diffraction , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Survival Rate
7.
J Radiat Res ; 52(4): 472-80, 2011.
Article in English | MEDLINE | ID: mdl-21785235

ABSTRACT

Fission-neutron radiation damage is hard to treat due to its critical injuries to hematopoietic and gastrointestinal systems, and so far few data are available on the therapeutic measures for neutron-radiation syndrome. This study was designed to test the effects of recombinant human granulocyte colony-stimulating factor (rhG-CSF) in dogs which had received 2.3 Gy mixed fission-neutron-γ irradiation with a high ratio of neutrons (~90%). Following irradiation, rhG-CSF treatment induced 100% survival versus 60% in controls. Only two of five rhG-CSF-treated dogs experienced leukopenia (white blood cells [WBC] count < 1.0 × 10(9)/L) and neutropenia (neutrophil [ANC] count < 0.5 × 10(9)/L), whereas all irradiated controls displayed a profound period of leukopenia and neutropenia. Furthermore, administration of rhG-CSF significantly delayed the onset of leukopenia and reduced the duration of leucopenia as compared with controls. In addition, individual dogs in the rhG-CSF-treated group exhibited evident differences in rhG-CSF responsiveness after neutron-irradiation. Finally, histopathological evaluation of the surviving dogs revealed that the incidence and severity of bone marrow, thymus and spleen damage decreased in rhG-CSF-treated dogs as compared with surviving controls. Thus, these results demonstrated that rhG-CSF administration enhanced recovery of myelopoiesis and survival after neutron-irradiation.


Subject(s)
Granulocyte Colony-Stimulating Factor/metabolism , Radiation Injuries, Experimental/drug therapy , Recombinant Proteins/metabolism , Animals , Cell Survival , Dogs , Gamma Rays , Humans , Leukopenia/drug therapy , Leukopenia/radiotherapy , Myeloid Cells/cytology , Neutrons , Neutropenia/drug therapy , Neutropenia/radiotherapy , Neutrophils/drug effects , Neutrophils/radiation effects , Whole-Body Irradiation
8.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 18(6): 1520-4, 2010 Dec.
Article in Chinese | MEDLINE | ID: mdl-21176363

ABSTRACT

The aim of this study was to investigate the effect of recombinant human granulocyte stimulating factor (rhG-CSF) on blood coagulation of beagles irradiated by 2.3 Gy neutron so as to provide new therapy for blood coagulation disorder after neutron irradiation. 10 beagles were exposed to 2.3 Gy neutron, and then randomly assigned into supportive care group and rhG-CSF-treated group. The rhG-CSF-treated cohorts were injected subcutaneously with rhG-CSF (10 µg/kg·d) beginning at the day of exposure for 21 consecutive days. Peripheral blood platelet counts were examined once every two days. In vitro platelet aggregation test, thromboelastography and blood clotting tetrachoric tests were also performed. The results indicated that the blood clotting system of irradiated dogs was in hypercoagulable state in the early days after 2.3 Gy neutron irradiation, and became hypocoagulable at crisis later and were mainly on intrinsic coagulation pathway. Blood fibrinogen increased markedly during the course of disease, while platelet counts and aggregation function were decreased remarkably. rhG-CSF administered daily could correct hypercoagulable state induced by 2.3 Gy neutron irradiation at the early time post exposure, shortened the thromboplastin generation time and clotting formation, down-regulated the abnormal high fibrinogen in blood, and improved platelet aggregation function. It is concluded that rhG-CSF can improve coagulation disorders of irradiated dogs.


Subject(s)
Blood Coagulation/drug effects , Bone Marrow/radiation effects , Granulocyte Colony-Stimulating Factor/pharmacology , Radiation Injuries, Experimental/physiopathology , Animals , Dogs , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Leukocyte Count , Neutron Diffraction , Platelet Count , Radiation Dosage , Recombinant Proteins
9.
J Pharm Pharmacol ; 62(4): 521-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20604843

ABSTRACT

OBJECTIVES: Total steroidal saponins extracted from the rhizome of Paris polyphylla (TSSP) have been used in China for the treatment of abnormal uterine bleeding. The aim of this study was to analyse the structure-activity relationship of steroidal saponins purified from P. polyphylla Sm. var. yunnanensis on rat myometrial contractions, and investigate the synergism among themselves as well as with known inherent agonists, such as Prostaglandin F(2alpha) (PGF-2alpha). METHODS: In this study, 22 steroidal saponins purified from TSSP were screened for their contractile activity in isolated uterine strips from estrogen-primed rats. KEY FINDINGS: It was shown that spirostanol glycosides exhibited inducible or inhibitory activity in rat uterine contraction based on the difference of their structures, which was not only attributed in part to the number, the length and the position of sugar side chains attached by a glycoside, but also related to the structure of the aglycone. Furthermore, synergistic actions were observed among pennogenin or diosgenin glycosides as well as with the known inherent agonist PGF-2alpha, indicating they may share, at least in part, similar pathways with PGF-2alpha in stimulating myometrial contractions. Finally, the contractile response of rat myometrium to spirostanol glycosides was significantly enhanced with advancing pregnancy. CONCLUSIONS: Together, these data support the possibility that some spirostanol glycosides may represent a new type of contractile agonist for the uterus and their synergism may be responsible for the therapeutic effect of TSSP on abnormal uterine bleeding.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Liliaceae/chemistry , Myometrium/drug effects , Phytosterols/pharmacology , Saponins/pharmacology , Animals , Dinoprost/metabolism , Diosgenin/pharmacology , Drug Synergism , Drugs, Chinese Herbal/chemistry , Estrogens/pharmacology , Female , Myometrium/physiology , Phytosterols/chemistry , Pregnancy , Rats , Rats, Wistar , Rhizome , Saponins/chemistry , Structure-Activity Relationship
10.
Int J Cancer ; 127(6): 1259-70, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20087863

ABSTRACT

STAT3 protein has an important role in oncogenesis and is a promising anticancer target. Herein, we demonstrate that a novel small molecule fluacrypyrim (FAPM) inhibits the growth of leukemia cells by a predominant G1 arrest with significant decrease of the protein and mRNA levels of cyclin D1. As cyclin D1 is transcriptionally regulated by STAT3, FAPM is then shown to markedly inhibit the STAT3 phosphorylation with marginal effect on the other signal transducers and activators of transcription, and without effect on phosphoinositide-3-kinase and mitogen-activated protein kinase pathways. Further analysis shows that FAPM significantly increases the protein tyrosine phosphatases (PTPs) activity in a dose-dependent manner, and the inhibition of PTP activation by sodium pervanadate reverses FAPM-induced suppression of STAT3 tyrosine phosphorylation, indicating an important role of PTP in the action of FAPM. Finally, FAPM treatment results in selective suppression of STAT3-mediated transcriptional activity and its downstream effectors, and subsequent induction of growth arrest and apoptosis in STAT3-dependent cancer cell lines. This study therefore identifies FAPM as a potent STAT3 activation inhibitor with possible therapeutic potential against malignancies with constitutive STAT3 activation.


Subject(s)
Acrylates/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Pyrimidines/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Base Sequence , Blotting, Western , Breast Neoplasms/pathology , Cyclin D/genetics , DNA Primers , Down-Regulation/drug effects , Female , Flow Cytometry , G1 Phase/drug effects , HL-60 Cells , Humans , K562 Cells , Leukemia/metabolism , Leukemia/pathology , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , STAT3 Transcription Factor/metabolism , Transcription, Genetic/drug effects
11.
Int J Cancer ; 121(9): 2084-2094, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17640057

ABSTRACT

Melissoidesin G (MOG) is a new diterpenoid purified from Isodon melissoides, a plant used in Chinese traditional medicine as antitumor and anti-inflammatory agents. In our study, MOG was shown to specifically inhibit the growth of human leukemia cell lines and primary acute myeloid leukemia (AML) blasts via induction of apoptosis, with the evidence of mitochondrial DeltaPsim loss, reactive oxygen species production, caspases activation and nuclear fragmentation. Furthermore, it was shown that thiol-containing antioxidants completely blocked MOG-induced mitochondrial DeltaPsim loss and subsequent cell apoptosis, while the inhibition of apoptosis by benzyloxy-carbonyl-Val-Ala-Asp-fluoromethylketone only partially attenuated mitochondrial DeltaPsim loss, indicating that MOG-induced redox imbalance is an early event upstream to mitochondrial DeltaPsim loss and caspase-3 activation. Consistently, it was found that MOG rapidly decreased the intracellular glutathione (GSH) content in a dose-dependent manner and the significance of GSH depletion in MOG-induced apoptosis was further supported by the protective effects of tert-butylhydroquinone (tBHQ) and the facilitative effects of DL-buthionine (S,R)-sulfoximine (BSO). Furthermore, it was showed that GSH depletion induced by MOG rendered some leukemia cell lines more sensitive to arsenic trioxide (As2O3), doxorubicin or cisplatin. Additionally, the synergistic apoptotic effects of MOG with As2O3 were detected in HL-60 and primary AML cells, but not in normal cells, suggesting the selective toxicity of their combination to the malignant cells. Together, we proposed that MOG alone or administered with other anticancer agents may provide a novel therapeutic strategy for leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Diterpenes/pharmacology , Isodon/chemistry , Leukemia/metabolism , Leukemia/pathology , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Arsenic Trioxide , Arsenicals/pharmacology , Caspases/metabolism , Cytochromes c/metabolism , Diterpenes/chemistry , Diterpenes/isolation & purification , Glutathione/metabolism , Humans , Mitochondria/drug effects , Molecular Structure , Oxidation-Reduction , Oxides/pharmacology , Phytotherapy , Tumor Cells, Cultured
12.
Am J Respir Cell Mol Biol ; 37(1): 9-19, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17413032

ABSTRACT

SARS-CoV 3a is a structural protein, mainly localizing to Golgi apparatus and co-localizing with SARS-CoV M in co-transfected cells. Here we observed that transient expression of 3a inhibited cell growth and prevented 5-bromodeoxyuridine incorporation, suggesting that 3a deregulated cell cycle progression. Cell cycle analysis demonstrated that 3a expression was associated with blockage of cell cycle progression at G1 phase in HEK 293, COS-7, and Vero cells 24-60 h after transfection. Mutation analysis of 3a revealed that C-terminal region (176 aa approximately 274 aa), including a potential calcium ATPase motif, was essential for induction of cell cycle arrest. Topological analysis showed that 3a predominantly located in Golgi apparatus, with its N-terminus residing in the lumen (Nlum) and C-terminus in the cytosol (Ccyt). Analyzing the cellular proteins involving in regulation of cell cycle progression, we demonstrated that 3a expression was correlated with a significant reduction of cyclin D3 level and phosphorylation of retinoblastoma (Rb) protein at Ser-795 and Ser-809/811, not with the expression of cyclin D1, D2, cdk4, and cdk6 in 293 cells. Increases in p53 phosphorylation on Ser-15 were observed in both SARS-CoV M and 3a transfected cells, suggesting that it might not correlate with the 3a-induced G0/G1 phase arrest. The reduction of cyclin D3 level and phosphorylation of Rb were further confirmed in SARS-CoV infected Vero cells. These results indicate that SARS-CoV 3a protein, through limiting the expression of cyclin D3, may inhibit Rb phosphorylation, which in turn leads to a block in the G1 phase of the cell cycle and an inhibition of cell proliferation.


Subject(s)
Cyclins/metabolism , G1 Phase , Retinoblastoma Protein/metabolism , Viral Proteins/physiology , Animals , COS Cells , Cell Line , Cell Proliferation , Chlorocebus aethiops , Cyclin D3 , Golgi Apparatus/metabolism , Humans , Models, Biological , Mutation , Protein Structure, Tertiary , Vero Cells , Viral Envelope Proteins , Viral Proteins/metabolism , Viroporin Proteins
13.
Mol Cells ; 21(2): 186-91, 2006 Apr 30.
Article in English | MEDLINE | ID: mdl-16682811

ABSTRACT

Severe acute respiratory syndrome-associated coronavirus (SARS-CoV), a distant member of the Group 2 coronaviruses, has recently been identified as the etiological agent of severe acute respiratory syndrome (SARS). The genome of SARS-CoV contains four structural genes that are homologous to genes found in other coronaviruses, as well as six subgroup-specific open reading frames (ORFs). ORF3 encodes a predicted 154-amino-acid protein that lacks similarity to any known protein, and is designated 3b in this article. We reported previously that SARS-CoV 3b is predominantly localized in the nucleolus, and induces G0/G1 arrest and apoptosis in transfected cells. In this study, we show that SARS-CoV 3b fused with EGFP at its N- or C- terminus co-localized with a mitochondria-specific marker in some transfected cells. Mutation analysis of SARS-CoV 3b revealed that the domain spanning amino acids 80 to 138 was essential for its mitochondria localization. These results provide new directions for studies of the role of SARS-CoV 3b protein in SARS pathogenesis.


Subject(s)
Mitochondria/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Fluorescent Dyes/metabolism , Humans , Mitochondria/genetics , Protein Sorting Signals , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Nonstructural Proteins/genetics
14.
Prep Biochem Biotechnol ; 36(3): 273-9, 2006.
Article in English | MEDLINE | ID: mdl-16707338

ABSTRACT

Lumbrokinase-3 (LK-3, AY438622), first cloned from the earthworm Eisenia foetida in our laboratory, is a component of earthworm fibrinolytic enzymes. In this study, cDNA encoding the LK-3 gene was sub-cloned into yeast pPIC9K expression vector and transformed into the Pichia pastoris GS115 cells by electroporation. High level expression of LK-3 in yeast cells was confirmed with a different induction time. The activity of expressed LK-3 was observed in fibrin plates. In addition, the expressed LK-3 protein could dissolve fibrinogen and bovine serum albumin. The use of this system for the high level production of biological protein is implicated from this study.


Subject(s)
Endopeptidases/biosynthesis , Endopeptidases/metabolism , Oligochaeta/enzymology , Pichia , Recombinant Proteins/biosynthesis , Animals , Endopeptidases/genetics , Gene Expression , Pichia/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Cytokine ; 33(4): 219-25, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16595180

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is the major cytokine involved in the control of neutrophil development. G-CSF activates the special receptor, the G-CSF receptor (GCSF-R), which subsequently triggers multiple signaling events. To obtain more interactive molecules with GCSF-R and to further understand the cellular signaling mechanism of GCSF-R, yeast two-hybrid system was used to screen a mouse liver library. Here, the interaction of GCSF-R and Snapin was found by yeast two-hybrid experiment, and the interaction of the two proteins was further confirmed by GST pull-down experiment, mammalian two-hybrid experiment and co-immunoprecipitation study. Moreover, the immuno-fluorescence assay was shown that the two proteins of GCSF-R with Snapin were co-localized in the cytoplasm and plasma membrane. The region of C-terminal GCSF-R between box2 and box3, including the residue Tyr703, was responsible for the interaction with Snapin. These data suggested that Snapin is a new interactive protein of GCSF-R.


Subject(s)
Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Signal Transduction/physiology , Vesicular Transport Proteins/metabolism , Animals , Cell Line , Humans , Mice , Protein Binding , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Two-Hybrid System Techniques , Vesicular Transport Proteins/genetics
16.
Virology ; 346(1): 74-85, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16303160

ABSTRACT

The genome of severe acute respiratory syndrome-associated coronavirus (SARS-CoV) contains four structural genes that are homologous to genes found in other coronaviruses, and also contains six subgroup-specific open reading frames (ORFs). Expression of one of these subgroup-specific genes, ORF7a, resulted in apoptosis via a caspase-dependent pathway. Here, we observed that transient expression of ORF7a protein fused with myc or GFP tags at its N or C terminus inhibited cell growth and prevented BrdU incorporation in different cultural cells, suggesting that ORF7a expression may regulate cell cycle progression. Analysis by flow cytometry demonstrated that ORF7a expression was associated with blockage of cell cycle progression at G0/G1 phase in HEK 293 cells after 24 to 60 h post-transfection. Similar results were observed in COS-7 and Vero cells. Mutation analysis of ORF7a revealed that the domain spanning aa 44-82 of 7a protein was essential for its cytoplasmic localization and for induction of the cell cycle arrest. After analyzing the cellular proteins involving in regulation of cell cycle progression, we demonstrated that ORF7a expression was correlated with a significant reduction of cyclin D3 level of mRNA transcription and expression, and phosphorylation of retinoblastoma (Rb) protein at ser795 and ser809/811, not with the expression of cyclin D1, D2, cdk4 and cdk6 in HEK 293 cells. These results suggest that the insufficient expression of cyclin D3 may cause a decreased activity of cyclin D/cdk4/6, resulting in the inhibition of Rb phosphorylation. Accumulation of hypo- or non-phosphorylated pRb thus prevents cell cycle progression at G0/G1 phase.


Subject(s)
Cyclins/metabolism , G1 Phase , Gene Expression Regulation , Membrane Proteins/metabolism , Resting Phase, Cell Cycle , Retinoblastoma Protein/metabolism , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Viral Proteins/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cyclin D3 , Cyclins/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mutation , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinoblastoma Protein/genetics , Severe acute respiratory syndrome-related coronavirus/physiology , Transcription, Genetic , Transfection , Vero Cells , Viral Matrix Proteins , Viral Proteins/chemistry , Viral Proteins/genetics
17.
Virol J ; 2: 66, 2005 Aug 17.
Article in English | MEDLINE | ID: mdl-16107218

ABSTRACT

Severe Acute Respiratory Syndrome coronavirus (SARS-CoV), cause of the life-threatening atypical pneumonia, infects many organs, such as lung, liver and immune organ, and induces parenchyma cells apoptosis and necrosis. The genome of SARS-CoV, not closely related to any of the previously characterized coronavirus, encodes replicase and four major structural proteins and a number of non-structural proteins. Published studies suggest that some non-structural proteins may play important roles in the replication, virulence and pathogenesis of viruses. Among the potential SARS-CoV non-structural proteins, 3b protein (ORF4) is predicted encoding 154 amino acids, lacking significant similarities to any known proteins. Till now, there is no report about the function of 3b protein. In this study, 3b gene was linked with the EGFP tag at the C- terminus. Through cell cycle analysis, it was found that over-expression of 3b-EGFP protein in Vero, 293 and COS-7 cells could induce cell cycle arrest at G0/G1 phase, and that especially in COS-7 cells, expression of 3b-EGFP was able to induce the increase of sub-G1 phase from 24 h after transfection, which was most obvious at 48 h. The apoptosis induction of 3b fusion protein in COS-7 cells was further confirmed by double cell labeling with 7-AAD and Annexin V, the function of 3b protein inducing cell G0/G1 arrest and apoptosis may provide a new insight for further study on the mechanism of SARS pathogenesis.


Subject(s)
Apoptosis , Cytokinesis/drug effects , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Viral Nonstructural Proteins/physiology , Animals , COS Cells , Chlorocebus aethiops , Vero Cells
18.
Virus Res ; 114(1-2): 70-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16046244

ABSTRACT

The open reading frame 3 (ORF3) of the severe acute respiratory syndrome coronavirus (SARS-CoV) genome encodes a predicted 154-amino acid protein, which lacks similarities to any known protein, and is named 3b. In this study, it was shown that 3b protein was predominately localized to nucleus with EGFP tag at its N- or C-terminus. The localization patterns were similar in different transfected cells. Immuno-fluorescence assay revealed that 3b protein was co-localized well with C23 in nucleolus. C23, B23 and fibrillarin all are important nucleolar proteins, which localize in the region of the nucleolus. Co-transfection of p3b-EGFP with pC23-DsRed, pB23-DsRed and pfibrillarin-DsRed further confirmed 3b's nucleolus localization. With construction of serial truncated mutants of 3b, a region (residues 134-154 aa) responsible for nucleolar localization was determinated in 3b protein. These results provide a new insight for further functional studies of SARS-CoV 3b protein.


Subject(s)
Cell Nucleolus/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Chromosomal Proteins, Non-Histone/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Nuclear Proteins/metabolism , Nucleophosmin , Phosphoproteins/metabolism , Protein Sorting Signals , RNA-Binding Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics , Transfection , Vero Cells , Viral Nonstructural Proteins/chemistry , Nucleolin
19.
Virus Res ; 109(2): 191-202, 2005 May.
Article in English | MEDLINE | ID: mdl-15763150

ABSTRACT

SARS-CoV 3a protein was a unique protein of SARS coronavirus (SARS-CoV), which was identified in SARS-CoV infected cells and SARS patients' specimen. Recent studies revealed that 3a could interact specifically with many SARS-CoV structural proteins, such as M, E and S protein. Expressed 3a protein was reported to localize to Golgi complex in SARS-CoV infected cells. In this study, it was shown that 3a protein was mainly located in Golgi apparatus with different tags at N- or C-terminus. The localization pattern was similar in different transfected cells. With the assay of truncated 3a protein, it was shown that 3a might contain three transmembrane regions, and the second or third region was properly responsible for Golgi localization. By ultra-centrifugation experiment with different extraction buffers, it was confirmed that 3a was an integral membrane protein and embedded in the phospholipid bilayer. Immunofluorescence assay indicated that 3a was co-localized with M protein in Golgi complex in co-transfected cells. These results provide a new insight for further study of the 3a protein on the pathogenesis of SARS-CoV.


Subject(s)
Severe acute respiratory syndrome-related coronavirus , Subcellular Fractions/chemistry , Viral Proteins/analysis , Animals , COS Cells , Cell Fractionation , Cell Line , Chlorocebus aethiops , Coronavirus M Proteins , Fluorescent Antibody Technique , Golgi Apparatus/chemistry , Humans , Intracellular Membranes/chemistry , Lipid Bilayers/chemistry , Membrane Proteins/analysis , Protein Structure, Tertiary , Transfection , Vero Cells , Viral Envelope Proteins , Viral Matrix Proteins/analysis , Viral Proteins/chemistry , Viroporin Proteins
20.
Acta Biochim Biophys Sin (Shanghai) ; 36(4): 303-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15253157

ABSTRACT

The earthworm fibrinolytic enzyme-3 (EFE-3, GenBank accession No: AY438622), from the earthworm Eisenia foetida, is a component of earthworm fibrinolytic enzymes. In this study, cDNA encoding the EFE-3 was cloned by RT-PCR. The cDNA contained an open reading frame of 741 nucleotides, which encoded a deduced protein of 247 amino acid residues, including signal sequences. EFE-3 showed a high degree of homology to earthworm (Lumbricus rebullus) proteases F-III-1, F-III-2, and bovine trypsin. The recombinant EFE-3 was expressed in E. coli as inclusion bodies, and the gene encoding the native form of EFE-3 was expressed in COS-7 cells in the medium. Both the refolding product of inclusion bodies and the secreted protease could dissolve the artificial fibrin plate.


Subject(s)
Cloning, Molecular , DNA, Complementary/genetics , Endopeptidases/chemistry , Endopeptidases/genetics , Oligochaeta/enzymology , Oligochaeta/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Computational Biology , Culture Media , Electrophoresis, Polyacrylamide Gel , Endopeptidases/metabolism , Escherichia coli/genetics , Fibrinolysis , Gene Expression , Inclusion Bodies/metabolism , Molecular Sequence Data , Open Reading Frames , Protein Folding , Protein Sorting Signals , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL