Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 881-899, 2021.
Article in English | WPRIM | ID: wpr-922771

ABSTRACT

The current study was designed to explore the brain protection mechanism of Xinglou Chengqi Decoction (XCD) based on gut microbiota analysis and network pharmacology. A transient middle cerebral artery occlusion (MCAO) model of mice was established, followed by behavioral evaluation, TTC and TUNEL staining. Additionally, to investigate the effects of gut microbiota on neurological function after stroke, C57BL/6 mice were treated with anti-biotic cocktails 14 days prior to ischemic stroke (IS) to deplete the gut microbiota. High-throughput 16S rDNA gene sequencing, metabonomics technique, and flow multifactor technology were used to analyze bacterial communities, SCFAs and inflammatory cytokines respectively. Finally, as a supplement, network pharmacology and molecular docking were applied to fully explore the multicomponent-multitarget-multichannel mechanism of XCD in treating IS, implicated in ADME screening, target identification, network analysis, functional annotation, and pathway enrichment analysis. We found that XCD effectively improved neurological function, relieved cerebral infarction and decreased the neuronal apoptosis. Moreover, XCD promoted the release of anti-inflammatory factor like IL-10, while down-regulating pro-inflammatory factors such as TNF-α, IL-17A, and IL-22. Furthermore, XCD significantly increased the levels of short chain fatty acids (SCFAs), especially butyric acid. The mechanism might be related to the regulation of SCFAs-producing bacteria like Verrucomicrobia and Akkermansia, and bacteria that regulate inflammation like Paraprevotella, Roseburia, Streptophyta and Enterococcu. Finally, in the network pharmacological analysis, 51 active compounds in XCD and 44 intersection targets of IS and XCD were selected. As a validation, components in XCD docked well with key targets. It was obviously that biological processes were mainly involved in the regulation of apoptotic process, inflammatory response, response to fatty acid, and regulation of establishment of endothelial barrier in GO enrichment. XCD can improve neurological function in experimental stroke mice, partly due to the regulation of gut microbiota. Besises, XCD has the characteristic of "multi-component, multi-target and multi-channel" in the treatment of IS revealed by network pharmacology and molecular docking.


Subject(s)
Animals , Mice , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome , Mice, Inbred C57BL , Molecular Docking Simulation , Network Pharmacology , Stroke/drug therapy
2.
Journal of Experimental Hematology ; (6): 607-611, 2011.
Article in Chinese | WPRIM | ID: wpr-313933

ABSTRACT

The aim of this study was to investigate the apoptosis-inducing effect of artemisinin derivative SM1044 on Kasumi-1 cells and its possible mechanism. Kasumi-1 cells were treated with different concentrations of SM1044, the cell viability was evaluated by MTT assay. Cell apoptosis and cell cycle progression were assessed by using flow cytometry with Annexin-V/PI double staining and flow cytometry with PI staining respectively. The expression of apoptosis-related proteins caspase 3, PARP and the fusion protein AML1-ETO were detected by Western blot. The results indicated that SM1044 inhibited cell growth of Kasumi-1 cells in time- and dose-dependent manners. After exposure of Kasumi-1 cells to 1 µmol/L SM1044 for 24 hours, the cell viability was decreased to 50%. IC(50) of SM1044 to Kasumi-1 cells at 48 hours was 0.17 ± 0.067 µmol/L. SM1044 induced cell apoptosis in a caspase-dependent manner, and the apoptotic rate of Kasumi-1 cells increased as SM1044 concentration increased. Flow cytometry with PI staining revealed that SM1044 induced cell cycle arrest, and the proportion of cells in G(0)/G(1) phase increased from 58.33 ± 4.46% to 71.75 ± 2.24% after exposure to 5 µmol/L SM1044 for 24 hours. Western blot showed that SM1044 increased the expression of apoptosis-related proteins cPARP and cleaved caspase 3 and also degraded the AML1-ETO fusion protein. It is concluded that SM1044 can inhibit the proliferation of Kasumi-1 cells, induce cell apoptosis which may be related to the increased level of cleaved PARP and cleaved caspase 3. SM1044 can also induce cell arrest in G(0)/G(1) phase. As the fusion protein AML1-ETO degrades obviously, it can be the potential target of SM1044 in Kasumi-1 cells.


Subject(s)
Humans , Antineoplastic Agents , Pharmacology , Apoptosis , Artemisinins , Pharmacology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Leukemia, Myeloid, Acute , Pathology
3.
Chinese Journal of Hematology ; (12): 603-606, 2008.
Article in Chinese | WPRIM | ID: wpr-239975

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effects of CDA-II alone or combined with cAMP on the retinoic acid (RA)-resistant acute promyelocytic leukemia (APL) cells.</p><p><b>METHODS</b>The RA-resistant cell line NB4-R2 was used as an in vitro model and treated with CDA-II alone or in combination with cAMP. Cell apoptosis was assessed by morphology observation, distribution of cellular DNA contents and sub-G1 cell population. The level of Bcl-2 was detected by flow cytometry, DNA "ladder" was detected by agarose-electrophoresis.</p><p><b>RESULTS</b>CDA-II could induce NB4-R2 cell apoptosis through decreasing the level of cellular anti-apoptotic protein Bcl-2. cAMP could significantly enhance the role of CDA-II. Bcl-2 positive cell rates decreased to (15.1 +/- 4.8)% and (7.3 +/- 2.9)% in NB4-R2 cells treated with 1 mg/ml CDA-II plus 100 micromol/L cAMP for 48 h and 72 h, respectively. While 100 micromol/L of cAMP could decrease Bcl-2 positive NB4-R2 cells from (92.0 +/- 0.6)% to (75.3 +/- 2.0)%.</p><p><b>CONCLUSIONS</b>CDA-II combined with cAMP could exert potent apoptotic effect on RA-resistant APL cells.</p>


Subject(s)
Animals , Humans , Apoptosis , CD11c Antigen , Metabolism , Cells, Cultured , Cyclic AMP , Pharmacology , Leukemia, Promyelocytic, Acute , Drug Therapy , Pathology , Peptides , Pharmacology , Phenylacetates , Pharmacology , Proto-Oncogene Proteins c-bcl-2 , Metabolism , Tretinoin , Pharmacology
4.
Journal of Experimental Hematology ; (6): 44-47, 2008.
Article in Chinese | WPRIM | ID: wpr-318719

ABSTRACT

This study was aimed to investigate the possible effects of cyclic adenosine monophosphate (cAMP) analogue 8-(4-chlorophenylthio) adenosine 3', 5'-cyclic monophosphate (8-CPT-cAMP) on the M(2b) subtype of acute myeloid leukemia (AML-M(2b)) cells. AML-M(2b) is characterized by the non-random chromosome translocation t (8; 21) (q22; q22), through which AML1 (acute myeloid leukemia 1) gene on chromosome 21 is fused with ETO (eight twenty-one) gene on chromosome 8, coding correspondent AML1-ETO fusion protein, which plays a crucial role in the leukemogenesis of AML-M(2b). The AML-M(2b) cell line Kasumi-1 cells were used as an in vitro model. The influences of 8-CPT-cAMP on the proliferation and differentiation of Kasumi-1 cells were evaluated according to cellular morphology, changes in cell surface antigen and cell cycle, as well as nitroblue-tetrazolium (NBT) assay. Meanwhile, semi-quantity RT-PCR and Western blot assay were used to detect the degradation of AML1-ETO fusion protein in Kasumi-1 cells before and after the treatment. The results showed that 8-CPT-cAMP (200 micromol/L) could significantly inhibit cell growth and induce differentiation of Kasumi-1 cells. However, it must be pointed out that 8-CPT-cAMP-induced differentiation in Kasumi-1 is not a typical terminal differentiation. Furthermore, 8-CPT-cAMP exerted little influence on the expression of AML1-ETO fusion gene and its product in Kasumi-1 cells. In conclusion, the 8-CPT-cAMP induced differentiation in Kasumi-1 cells. This results may provide experimental and theoretical basis for the breakthrough of differentiation-induced therapy extended to another leukemia.


Subject(s)
Humans , Cell Transformation, Neoplastic , Core Binding Factor Alpha 2 Subunit , Genetics , Metabolism , Cyclic AMP , Pharmacology , Leukemia, Myeloid, Acute , Genetics , Metabolism , Pathology , Oncogene Proteins, Fusion , Genetics , Metabolism , RUNX1 Translocation Partner 1 Protein , Thionucleotides , Pharmacology , Tumor Cells, Cultured
5.
Chinese Journal of Integrated Traditional and Western Medicine ; (12): 909-911, 2007.
Article in Chinese | WPRIM | ID: wpr-245610

ABSTRACT

<p><b>OBJECTIVE</b>To understand the effects of Chinese herbal medicine for stasis -removing (SR) and muscle-regenerating (MR), in different constitutions, on granulation tissue of ordinary wound.</p><p><b>METHODS</b>Shengji Huayu recipe, a common used Chinese recipe for SR & MR (SR-MR), and its disassembled recipe, Shengji recipe (MR) and Huayu recipe (SR), were applied resectively on the full-layer skin injured wound in 120 rats to observe their effects on collagen type I (C I ) and matrix metalloproteinase-1 (MMP-1) in granulation tissue of wound at various stages of healing with immunohistochemistry technique and image-analysis system.</p><p><b>RESULTS</b>The expression of C I of the normal group was obviously delayed and the level of MMP-1 increased in a ladder type from day 3 to day 11. Shengji group could promote the secretion of C I and MMP-1 at the early stage and keep a high level from day 3 to day 11. The expression of MMP-1 of Huayu group maintained a high level on the first 7 days with significant difference as compared with the Shengji group (P < 0.05), and reduced from day 11 showing significant difference when compared with the normal group and Shengji group (all P < 0.05). Shengji Huayu low dose group has two periods of peak time in promoting the secretion of C I on day 7 and 15, and showed significant difference as compared with the normal group (P < 0.05); Shengji Huayu high dose group could reduce the MMP-1 on day 11 and obviously lower when compared with the normal group (P<0.05).</p><p><b>CONCLUSION</b>The mechanism of Chinese herbal medicine for SR-MR in promoting wound healing was probably through inhibiting the secretion of MMP-1 to increase the C I content in granulation tissue of wound.</p>


Subject(s)
Animals , Male , Rats , Collagen Type I , Drugs, Chinese Herbal , Therapeutic Uses , Granulation Tissue , Metabolism , Pathology , Immunohistochemistry , Matrix Metalloproteinase 1 , Medicine, Chinese Traditional , Methods , Phytotherapy , Rats, Sprague-Dawley , Wound Healing , Wounds and Injuries , Drug Therapy , Metabolism
6.
Chinese Journal of Hematology ; (12): 150-153, 2006.
Article in Chinese | WPRIM | ID: wpr-243981

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the mechanism of clinical haemorrhage in an inherited coagulation factor VII (FVII) deficiency and tissue factor abnormality pedigree.</p><p><b>METHODS</b>All exons, exon-intron boundaries and the 3', 5' untranslated sequences of FVII and tissue factor (TF) genes were amplified by PCR and sequenced directly. Any mutation identified by direct sequencing was confirmed by reverse sequencing. FVII cDNA of the proband was synthesized with random primers and amplified by nest PCR.</p><p><b>RESULTS</b>55C-->T heterozygous mutation located in promoter of FVII gene was identified in the proband. The heterozygous mutation was derived from his mother. Tracing the other pedigree members found that his sister had the same heterozygous mutation and the others had wild-type FVII genes. A 9363 C-->T (Arg131Trp) heterozygous polymorphism in TF gene, which was 2.63% frequency of T allele polymorphism, was found in all of the pedigree members.</p><p><b>CONCLUSION</b>It was the first report that the -55C-->T heterozygous mutation in FVII gene and the Arg131Trp heterozygous polymorphism in TF gene explained the clinical symptom of the proband.</p>


Subject(s)
Adult , Humans , Male , DNA Mutational Analysis , Factor VII , Genetics , Factor VII Deficiency , Genetics , Heterozygote , Pedigree , Polymorphism, Genetic , Thromboplastin , Genetics
7.
Journal of Experimental Hematology ; (6): 1086-1089, 2005.
Article in Chinese | WPRIM | ID: wpr-343821

ABSTRACT

Congenital afibrinogenemia is a rare autosomal recessive disorder, characterized by the complete absence or extremely reduced level of fibrinogen. To analyze the phenotype and genotype of a family with inherited afibrinogenemia, laboratory studies including activated partial thromboplastin time (APTT), prothrombin time (PT) and thrombin time (TT) were tested in the proband and 9 family members. Fibrinogen (Fg) in plasma were measured by both functional and immunoturbidimetry assay. All the exons, exon-intron boundaries and promoter regions of three Fg genes were analyzed by direct sequencing. 102 healthy blood donors were used as normal control. The results showed that phenotype of the proband was diagnosed as afibrinogenemia. Compound heterozygous mutations in Fg FGB gene were detected in the proband. One was a nonsense mutation (Arg17stop) in exon 2, traced back to the proband's mother. The other was a missense mutation (Gly347Arg) in exon 7, which was from the proband' s father. It is concluded that afibrinogenemia is caused by the compound heterozygous mutations Arg17stop and Gly347Arg in the Beta beta-chain of fibrinogen.


Subject(s)
Adult , Child , Female , Humans , Male , Middle Aged , Afibrinogenemia , Genetics , Amino Acid Sequence , Base Sequence , Codon, Nonsense , DNA Mutational Analysis , Fibrinogen , Chemistry , Genetics , Heterozygote , Molecular Sequence Data , Mutation, Missense , Pedigree , Phenotype , Protein Structure, Secondary , Sequence Homology, Amino Acid
8.
Chinese Journal of Hematology ; (12): 144-147, 2005.
Article in Chinese | WPRIM | ID: wpr-229881

ABSTRACT

<p><b>OBJECTIVE</b>To identify gene defect in a Chinese pedigree of hereditary coagulation factor XI (FXI) deficiency.</p><p><b>METHODS</b>The peripheral blood samples were collected from the proband and her family members. The plasma PT, APTT, FXI:C and FXI:Ag were assayed. The FXI gene exons and exon-intron boundaries of the proband were amplified by PCR and then sequenced directly. The mRNA of FXI in the peripheral blood was analyzed with RT-PCR.</p><p><b>RESULTS</b>The proband and some of her family members had prolonged APTT. The plasma FXI:C and FXI:Ag of the proband, her brother and her parents were lower than 10% and 50% of the normal values, respectively. Nucleotide sequence analysis revealed that the proband and her brother had a homozygous mutation of IVS J-4delgttg in FXI gene. The mutation was inherited from her parents who were heterozygotes. The mutation was not found in 60 normal subjects. No FXI mRNA was detected in peripheral blood sample of the proband.</p><p><b>CONCLUSION</b>The IVS J-4delgttg is a novel mutation causing FXI deficiency, which may interfere with mRNA splicing.</p>


Subject(s)
Adult , Female , Humans , Base Sequence , DNA Mutational Analysis , Factor XI , Genetics , Factor XI Deficiency , Blood , Genetics , Pathology , Genotype , Introns , Genetics , Molecular Sequence Data , Partial Thromboplastin Time , Pedigree , Phenotype , Point Mutation , Prothrombin Time , RNA, Messenger , Genetics , Metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion
9.
Chinese Journal of Hematology ; (12): 148-151, 2005.
Article in Chinese | WPRIM | ID: wpr-229880

ABSTRACT

<p><b>OBJECTIVE</b>To study the molecular mechanism of antithrombin (AT) gene C2759T (Leu99Phe) mutation causing AT deficiency.</p><p><b>METHODS</b>A mutated AT cDNA expression plasmid ATM2759 was constructed by mega-primer method. ATM2759 and wild type AT cDNA expression plasmid ATN were transfected into COS7 cells or CHO cells by using Superfect reagent respectively for in vitro expression study and immunofluorescence assay.</p><p><b>RESULTS</b>The antigen levels of AT (AT:Ag) in the cell lysate of ATM2759 transfected COS7 cells and the cell culture supernatant were 174.97% and 35.63% of that of ATN transfected COS7 cells respectively, whereas the AT activity in the cell culture supernatant was 47.73% of the control's. Immunofluorescence analysis showed that the fluorescence intensity was significantly higher in ATM2759 transfected CHO cells than in those transfected with ATN.</p><p><b>CONCLUSIONS</b>Leu99Phe substitution may not affect the binding capacity of AT with heparin. Secretion defect and intracellular accumulation of the mutated AT protein might be the mechanisms of this mutation causing AT deficiency.</p>


Subject(s)
Animals , Cricetinae , Antithrombin III , Genetics , Metabolism , Antithrombin III Deficiency , Genetics , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetulus , Fluorescent Antibody Technique , Mutation , Plasmids , Genetics , Transfection
10.
Journal of Experimental Hematology ; (6): 169-173, 2005.
Article in Chinese | WPRIM | ID: wpr-347802

ABSTRACT

One of the important approaches for further prolonging remission duration and eradicating minimal residual disease in acute leukemia is immunotherapy. Four kinds of immunotherapy for acute leukemia are under investigation: (1) monoclonal antibodies, among them, Mylotarg (cytotoxic antibiotic calicheamicin linked to CD33 Mab) is given for the treatment of refractory or relapsed acute myeloid leukemia and molecular relapse in acute promyelocytic leukemia with good results, Campath-1H (antiCD52 Mab) is administered in the treatment of prolymphocytic leukemia and Rituximab (anti-CD20 Mab) in B-PLL with high complete remission rates. Other Mabs under preclinical and clinical trials include anti-IL-2 receptor Mab for the treatment of acute T lymphocytic leukemia, anti-220 kD Mab-6G7 for acute leukemias, recombinant immune toxin BL22 (anti-CD22) for hairy cell leukemia and Mabs labeled with radio-isotopes for different types of acute leukemias; (2) adoptive cellular immunotherapy using cytokine-induced killer cell, alloreactive NK cells, allogeneic or autologous leukemic-specific CD8(+) cytotoxic T lymphocytes, and other immune effector cells; (3) cytokines and other immune modulators comprising IL-2, IL-12, GM-CSF, CD40L, FLT-3L and thalidomide and its derivatives; (4) leukemia vaccines of several different formulations including antigen-specific, leukemia cell-based, leukemia antigen-pulsed dendritic cell (DC) and leukemia-derived DC vaccines, the latter two formulations are more attractive. In conclusion, up to now, the most effective example of immunotherapy in acute leukemia is provided by the administration of Mabs, and the majority of other approaches in immunotherapy for acute leukemia although promising, need further studies.


Subject(s)
Humans , Acute Disease , Adoptive Transfer , Methods , Antibodies, Monoclonal , Allergy and Immunology , Therapeutic Uses , Cancer Vaccines , Therapeutic Uses , Immunotherapy , Methods , Leukemia , Allergy and Immunology , Therapeutics
11.
Journal of Experimental Hematology ; (6): 188-193, 2004.
Article in Chinese | WPRIM | ID: wpr-352101

ABSTRACT

The aim is to observe the expression of human factor VIII gene in mice tranduced in vivo and ex vivo. The vector pLNC-FVIII BD was generated by cloning a B-domain-deleted (760aa-1639aa) FVIII cDNA (FVIIIBD cDNA) into retroviral vector pLNCX. 2 x 10(6) of mouse bone marrow stroma cells transduced by LNC-FVIII BD were infused into 4-week-old BALB/c mice by tail-vein injection. pLNC-FVIII BD was conjugated with PAMAM dendrimer to form complex PAMAM-pLNC-FVIII BD, with which C57BL/6J were injected by tail vein (200 micro l contained 15 micro g/mouse) and sacrificed at days 1, 2, 7, 14, 21 and 28, respectively after injection. Tissue such as liver, spleen, lung and kindney were harvested, with which the transcription were detected by means of RT-PCR. In addition, blood was collected to be measured human FVIII Ag, human FVIIIc and anti-FVIII of human inhibitors. The results showed that the highest level of human FVIII in the recipient BALB/c mice was 8.6 +/- 1.44 ng/ml detected on the first day post-injection; anti-FVIII antibodies were detected from the first week post-injection, and then the level of FVIII Ag decreased and cannot be measured on the fourth week. In the C57BL/6J mice physiological level of human FVIII was expressed in plasma at 48 hours after injection and the average human FVIIIc was 0.62 U/ml and the average human FVIII Ag was 115.5 ng/ml, and gradually reduced later. Anti-FVIII of human inhibitors was not revealed all the time. Syngene image scanning demonstrated that the transcription of the human FVIII BD cDNA occurred mainly in spleen and lung, and secondarily in liver and kidney. No side effects of PAMAM-pLNC-FVIII BD were observed in mice tissue by pathological examination at 4 weeks. In conclusion, retrovirus-transduced bone marrow stroma cells effectively produced human FVIII after ex vivo transduction, but the development of anti-FVIII antibodies in recipient mice influenced the expression level. The human FVIII gene can successfully be transduced in vivo through injecting PAMAM-pLNC-FVIII BD cDNA into mice intravenously. There was physiological level expression of human FVIII in plasma at 48 hours after injection and the average human FVIIIc is 0.62 U/ml and the peak in the six mice was 0.89 U/ml, and gradually reduced later.


Subject(s)
Animals , Mice , DNA, Complementary , Factor VIII , Genetics , Genetic Therapy , Hemophilia A , Therapeutics , Mice, Inbred BALB C , Mice, Inbred C57BL , Transfection
12.
Chinese Journal of Hematology ; (12): 675-678, 2004.
Article in Chinese | WPRIM | ID: wpr-229928

ABSTRACT

<p><b>OBJECTIVE</b>To explore the molecular mechanism of APL cell resistance to ATRA.</p><p><b>METHODS</b>The ATRA sensitive and resistant APL cell lines, NB4 and NB4-R1, were used as in vitro models. The effects of specific inhibitors and activators of adenylate cyclase (AC) and phosphodiesterase (PDE) on ATRA-induced differentiation was evaluated by cell morphology, cell surface antigen expression and nitroblue-tetrazolium (NBT) reduction assays.</p><p><b>RESULTS</b>SQ22536, a specific antagonist of AC, could dramatically block ATRA-induced NB4 cell differentiation. When ATRA + SQ22536 group compared with ATRA group, the positivity of CD11b decreased from (95.9 +/- 2.5)% to (60.3 +/- 7.1)%, while the A(540) in NBT reduction assay decreased from 0.585 +/- 0.092 to 0.170 +/- 0.028 (P < 0.05). Forskolin, an agonist of AC, could overcome the resistance of NB4-R1 cells to ATRA. When ATRA + forskolin group compared with ATRA group, the positivity of CD11b increased from (34.3 +/- 5.3)% to (94.6 +/- 2.4)%, while the A(540) in NBT reduction assay increased from 0.110 +/- 0.028 to 0.395 +/- 0.049 (P < 0.05). In contrast, the specific antagonist and agonist of PDE, 3-isobutyl-1-methylxanthine (IBMX) and calmodulin, exerted little impact on ATRA treatment.</p><p><b>CONCLUSIONS</b>The defaults in the initiation of AC activation may contribute to the resistance to ATRA in some APL cells.</p>


Subject(s)
Humans , Adenine , Pharmacology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases , Metabolism , Antineoplastic Agents , Pharmacology , CD11b Antigen , Metabolism , Cell Differentiation , Cell Line, Tumor , Drug Resistance, Neoplasm , Enzyme Activation , Enzyme Inhibitors , Pharmacology , Leukemia, Promyelocytic, Acute , Metabolism , Pathology , Phosphoric Diester Hydrolases , Metabolism , Tretinoin , Pharmacology
13.
Chinese Medical Journal ; (24): 71-74, 2004.
Article in English | WPRIM | ID: wpr-235830

ABSTRACT

<p><b>BACKGROUND</b>Coagulation factor VII (FVII) levels in plasma are usually related to ischemic heart disease (IHD) and cerebral infarction shares many of the risk factors related to IHD. Is there any relationship between factor VII and cerebral infarction? We investigated the relationship between FVII and acute cerebral infarction and reported genotype frequencies and allelic frequencies of FVII gene polymorphisms in the Chinese Han population.</p><p><b>METHODS</b>We recruited 62 patients with acute cerebral infarction confirmed by magnetic resonance imaging (MRI) from Ruijin Hospital, and 149 age-matched patients clinically free of vascular disease to act as controls. All of them were unrelated, and were from the Chinese Han population. FVII coagulant activity (FVIIc) was determined using an clotting assay, activated FVII (FVIIa) and FVII Ag were assayed using enzyme immunoassay kits. The FVII gene polymorphisms to be detected included-401G/T, -402G/A, 5'F7A1/A2, IVS7 and R353Q. 5'F7 and IVS7 were revealed by means of a PCR and direct agarose gel electrophoresis. The rest were examined by a polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP).</p><p><b>RESULTS</b>The results showed that FVIIc, FVIIAg and FVIIa were higher in the acute cerebral infarction group than in the control group (P < 0.01, P < 0.05, P < 0.05, respectively). There were no significant differences in the genotype frequencies of FVII gene polymorphisms between the two groups. The allelic frequencies in the Chinese Han population were as follows: -401G/T (96.64/3.36), -402G/A (52.01/47.99), 5'F7A1/A2 (96.64/3.36), IVS7 H5/H6/H7/H8 (0.34/52.35/46.98/0.34) and R353Q (95.64/4.36). There were significant differences (P < 0.01, P < 0.001, P < 0.001, P < 0.001, P < 0.001, respectively) in these allelic frequencies between the Chinese Han and European populations.</p><p><b>CONCLUSIONS</b>The results indicate that increased plasma FVII levels may contribute to thrombosis in cerebral infarction. And there was no significant difference in genotype frequencies of these five FVII gene polymorphisms between the acute cerebral infarction and control groups. Moreover, these results showed that the frequencies of protective allele, including -401T, 5'F7 A2 and 353Q were lower, but that -402A, which was previously found to be associated with increased plasma FVII levels, is higher in Chinese Han population.</p>


Subject(s)
Humans , Acute Disease , Asian People , Genetics , Cerebral Infarction , Genetics , China , Europe , White People , Genetics , Factor VII , Genetics , Gene Frequency , Intracranial Thrombosis , Genetics , Polymorphism, Genetic
14.
Chinese Medical Journal ; (24): 813-817, 2004.
Article in English | WPRIM | ID: wpr-284901

ABSTRACT

<p><b>BACKGROUND</b>We identified the gene mutations in two Chinese pedigree of type I hereditary protein C deficiency and type I hereditary antithrombin deficiency.</p><p><b>METHODS</b>The plasma level of protein C activity (PC:A), protein C antigen (PC:Ag), protein S activity, antithrombin activity (AT:A) and antithrombin antigen (AT:Ag) of propositi and two family members were detected using ELISA and chromogenic assay, respectively. All exons and intron-exon boundaries of protein C gene and antithrombin gene were analyzed by direct sequencing of the corresponding amplified PCR products in DNA from the propositus.</p><p><b>RESULTS</b>The plasma PC:A and PC:Ag of propositus 1 was 26% and 1.43 mg/dl, respectively. The PC:Ag and PC:A of his father were normal. The decreased PC:A level was seen in his mother and 4 of his maternal pedigree. PS:A and AT:A were all normal in pedigree 1 members. A C5498T heterozygous mutation in exon 3 of protein C gene, resulting in the substitution of Arg for Trp at the 15th amino acid, was identified in propositus 1 and 8 of his relatives. The plasma AT:A and AT:Ag of propositus 2 was 48.6% and 10.4 mg/dl, respectively. The reduced AT:A and AT:Ag levels were found in his father and 5 of paternal pedigree. PC:A, PC:Ag and PS:A were all in normal range. A heterozygous 13387-9G deletion in exon 6 of antithrombin gene was identified in propositus 2. This mutation introduced a frameshift and a premature stop at codon 426 and existed in 6 members of pedigree 2.</p><p><b>CONCLUSION</b>The C5498T heterozygous mutation in exon 3 of protein C gene, first reported in China, leads to type I hereditary protein C deficiency. The 13387-9G deletion, a novel mutation, can cause antithrombin deficiency and thrombosis.</p>


Subject(s)
Adolescent , Child , Female , Humans , Male , Fibrin , Gene Deletion , Pedigree , Protein C , Genetics , Protein C Deficiency , Genetics
15.
Chinese Journal of Hematology ; (12): 262-265, 2004.
Article in Chinese | WPRIM | ID: wpr-291453

ABSTRACT

<p><b>OBJECTIVE</b>In order to investigate the leukemogenic potential of NUP98-PMX1 fusion gene in vivo.</p><p><b>METHODS</b>NUP98-PMX1 transgenic mice were generated, in which the fusion gene was driven by hCG promoter and expressed in myeloid cells at early stage of differentiation. Molecular cloning technology was used to construct NUP98-PMX1 transgenic plasmid. The genotype and phenotype of the NUP98-PMX1 transgenic mice were analyzed by PCR, RT-PCR, peripheral blood count (PBC), bone marrow (BM) cells morphology and pathological examination.</p><p><b>RESULTS</b>NIH3T3 cells transfected with NUP98-PMX1 fusion gene grew faster, formed colonies in soft agar, and developed tumors in 10 inoculated nude mice. Among 8 disordered NUP98-PMX1 transgenic mice, 4 developed myeloid leukemia-like phenotype, including 3 resembling human chronic myeloid leukemia.</p><p><b>CONCLUSION</b>NUP98-PMX1 has oncogenic activity and plays a crucial role in leukemogenesis.</p>


Subject(s)
Animals , Female , Humans , Male , Mice , Bone Marrow Cells , Metabolism , Pathology , Disease Models, Animal , Flow Cytometry , Gene Expression Regulation, Leukemic , Green Fluorescent Proteins , Genetics , Metabolism , Leukemia, Myeloid , Genetics , Pathology , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Nude , Mice, Transgenic , NIH 3T3 Cells , Nuclear Pore Complex Proteins , Genetics , Metabolism , Oncogene Proteins, Fusion , Genetics , Metabolism , Phenotype , Plasmids , Recombinant Fusion Proteins , Genetics , Metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
16.
Chinese Journal of Hematology ; (12): 132-135, 2004.
Article in Chinese | WPRIM | ID: wpr-291433

ABSTRACT

<p><b>OBJECTIVES</b>To identify the FXI gene mutations in two Chinese pedigrees of congenital factor XI deficiency.</p><p><b>METHODS</b>The peripheral blood samples were collected from the probands and their family members and the plasma FXI:C and FXI:Ag were determined. All the exons and exon-intron boundries of FXI gene were amplified with PCR and sequenced thereafter.</p><p><b>RESULTS</b>A nonsense mutation Trp228stop and two missense mutations Glu323Lys and Leu172Pro were disclosed in the two pedigrees. All mutations existed in a heterozygous state.</p><p><b>CONCLUSION</b>The FXI gene mutations Trp228stop, Glu323Lys and Leu172Pro attribute to the pathogenesis of the congenital factor XI deficiency in Chinese. The Leu172Pro is identified for the first time.</p>


Subject(s)
Adult , Child , Humans , Male , Middle Aged , Asian People , Genetics , Base Sequence , Factor XI , Genetics , Factor XI Deficiency , Genetics , Molecular Sequence Data , Mutation , Pedigree
17.
Chinese Journal of Hematology ; (12): 139-142, 2004.
Article in Chinese | WPRIM | ID: wpr-291431

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the pathogenesis of inherited coagulation factor VII (FVII) deficiency.</p><p><b>METHODS</b>The diagnosis was validated by coagulant parameter assay. FVII gene mutations were analysed in the proband by DNA direct sequencing of PCR products of all exons, exon-intron boundaries and the 3', 5'untranslated sequences. The mutations were confirmed by reverse sequencing. The ectopic transcripts of RT-PCR were used to confirm the characteristics of the mutation in non-canonical splice site (IVS1a + 5g > a).</p><p><b>RESULTS</b>Double heterozygous mutations in the propositus were identified: a T to G mutation at position 10961, resulting in His348Gln substitution, a non-canonical splice site (IVS1a + 5g > a) mutation, causing the new model of splice and frameshift mutation.</p><p><b>CONCLUSION</b>Double heterozygous mutations of His348Gln and IVS1a + 5g > a were identified in a propositus, the splicing pattern of the IVS1a + 5g > a mutation was reported for the first time.</p>


Subject(s)
Humans , Amino Acid Sequence , Base Sequence , Factor VII , Genetics , Factor VII Deficiency , Genetics , Heterozygote , Molecular Sequence Data , Mutation, Missense , RNA Splicing
18.
Chinese Journal of Hematology ; (12): 519-522, 2004.
Article in Chinese | WPRIM | ID: wpr-291388

ABSTRACT

<p><b>OBJECTIVE</b>To explore the molecular mechanisms involved in a pedigree with inherited coagulation factor X (FX) deficiency.</p><p><b>METHODS</b>The activated partial thromboplastin time (APTT), prothrombin time (PT), FX activity (FX:C) and FX antigen (FX:Ag) test were adopted for phenotype diagnosis. All the 8 exons, intron/exon boundaries and the 5'untranslated regions (UTR) of the FX gene were amplified by polymerase chain reaction (PCR) from the genomic DNA extracted from the peripheral blood of the propositus. The PCR products were screened by direct sequencing. The mutation was confirmed by allele specific PCR (ASPCR).</p><p><b>RESULTS</b>The phenotype of the propositus was identified as FX deficiency (type II). Two novel FX gene mutations were detected in the propositus: one was a donor site splice mutation in intron 1 (IVS1 + 1G-->A), another was a missense mutation 1185G-->A in exon 8 (Arg347His).</p><p><b>CONCLUSION</b>The FX deficiency of the propositus is caused by double heterozygous mutations IVS1 + 1G-->A and Arg347His.</p>


Subject(s)
Female , Humans , Male , Young Adult , Antigens , Genetics , Base Sequence , DNA Mutational Analysis , Factor X , Genetics , Factor X Deficiency , Genetics , Heterozygote , Mutation , Pedigree
19.
Journal of Experimental Hematology ; (6): 721-725, 2004.
Article in Chinese | WPRIM | ID: wpr-347876

ABSTRACT

To investigate the non-viral vector mediating human coagulation factor VIII gene expression in mouse 32D cell line, a recombinant plasmid vector, pRC/RSV-hFVIIIBDcDNA, was constructed by cloning B-domain-deleted (Delta760aa-1639aa) human factor VIII cDNA (hFVIIIBDcDNA) into plasmid vector, pRC/RSV. The plasmid RC/RSV-hFVIIIBDcDNA was then transfected by means of SuperFect Transfection Reagent into mouse 32D cell line. After screening with G418, the procoagulant activity (hFVIII:C) and antigen (hFVIII:Ag) of human factor VIII in the culture medium were detected using one-stage method and ELISA, respectively. Furthermore, RT-PCR was performed to observe the transcription of hFVIIIBDcDNA. The results showed that human coagulation factor VIII protein existed in culture medium with hFVIII:C up to 2.01 U/(10(6) cell x 24 hours) and hFVIII:Ag to 450.08 ng/(10(6) cell x 24 hours). RT-PCR displayed mRNA of hFVIIIBDcDNA in 32D cells. It is concluded that the recombinant plasmid RC/RSV-hFVIIIBDcDNA can successfully express human FVIII in mouse 32D cell line, and hFVIII expressed in vitro presents the similar coagulant activity to the native hFVIII existing in normal human plasma.


Subject(s)
Animals , Humans , Mice , Cell Line , DNA, Complementary , Genetics , Enzyme-Linked Immunosorbent Assay , Factor VIII , Genetics , Metabolism , Gene Expression Regulation , Plasmids , Genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
20.
Chinese Journal of Medical Genetics ; (6): 279-283, 2003.
Article in Chinese | WPRIM | ID: wpr-248440

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the genotypes of mutations of an inherited coagulation factor VII(F VII) deficiency pedigree.</p><p><b>METHODS</b>The diagnosis was validated by coagulant parameters. F VII gene mutations were analysed in the proband and her family members by DNA direct sequencing. The PCR fragments were cleaved by the Msp I restriction enzyme to confirm the mutations detected by sequencing was performed in this study.</p><p><b>RESULTS</b>Double heterozygous mutations at the same coding site of amino acid were detected in propositus of the pedigree: a C to T mutation at position 11348 resulting in Arg304Trp substitution combined with a G to A mutation at position 11349 resulting in Arg304Gln substitution. Her farther had a G to A mutation at position 11349 and her mother had a C to T mutation at position 11348, respectively. Both were heterozygous mutations. One of her brothers had normal genotype, the other brother and all her three offsprings had heterozygous mutations.</p><p><b>CONCLUSION</b>Double heterozygous mutations coding the same amino acid were found in a pedigree with hereditary coagulation factor VII deficiency.</p>


Subject(s)
Female , Humans , Male , DNA Mutational Analysis , Factor VII , Genetics , Factor VII Deficiency , Genetics , Heterozygote , Mutation , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL