Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
J Gen Virol ; 101(12): 1242-1250, 2020 12.
Article in English | MEDLINE | ID: mdl-33016861

ABSTRACT

Post-translational modification plays a critical role in viral replication. Previously we reported that neddylation of PB2 of influenza A virus (IAV) can inhibit viral replication. However, we found that NEDD8 overexpression can still inhibit the replication of PB2 K699R mutant viruses, implying that other viral protein(s) can be neddylated. In this study, we revealed that M1 of IAV can also be modified by NEDD8. We found that the E3 ligase HDM2 significantly promotes M1 neddylation. Furthermore, we identified M1 K187 as the major neddylation site. We generated an IAV M1 K187R mutant (WSN-M1 K187R) and compared the growth of wild-type and mutant viruses in Madin-Darby canine kidney (MDCK) cells. The data showed that the replication of WSN-M1 K187R was more efficient than that of wild-type WSN. More importantly, we observed that overexpression of NEDD8 inhibited the replication of the wild-type WSN more effectively than that of WSN-M1 K187R. In addition, we found that the neddylation-deficient M1 mutant (M1 K187R) had a longer half-life than that of wild-type M1, indicating that the neddylation of M1 reduces stability. Then we performed a viral infection assay and found that WSN-M1 K187R exhibited greater virulence in mice than wild-type WSN, suggesting that the neddylation of M1 reduced IAV replication in vivo. In conclusion, we uncovered that neddylation of M1 by HDM2 negatively regulates the stability of M1, which in turn inhibits viral replication.


Subject(s)
Influenza A virus/physiology , NEDD8 Protein/metabolism , Orthomyxoviridae Infections/virology , Viral Matrix Proteins/metabolism , Virus Replication , Animals , Cell Line , Female , Humans , Influenza A virus/genetics , Influenza A virus/metabolism , Influenza A virus/pathogenicity , Lysine/genetics , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Protein Stability , Ubiquitin-Protein Ligases/metabolism , Viral Matrix Proteins/genetics , Virulence
2.
Korean J Parasitol ; 57(2): 101-115, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31104402

ABSTRACT

The pathogenesis of cerebral malaria is biologically complex and involves multi-factorial mechanisms such as microvascular congestion, immunopathology by the pro-inflammatory cytokine and endothelial dysfunction. Recent data have suggested that a pleiotropic T-cell immunomodulatory protein (TIP) could effectively mediate inflammatory cytokines of mammalian immune response against acute graft-versus-host disease in animal models. In this study, we identified a conserved homologue of TIP in Plasmodium berghei (PbTIP) as a membrane protein in Plasmodium asexual stage. Compared with PBS control group, the pathology of experimental cerebral malaria (ECM) in rPbTIP intravenous injection (i.v.) group was alleviated by the downregulation of pro-inflammatory responses, and rPbTIP i.v. group elicited an expansion of regulatory T-cell response. Therefore, rPbTIP i.v. group displayed less severe brain pathology and feverish mice in rPbTIP i.v. group died from ECM. This study suggested that PbTIP may be a novel promising target to alleviate the severity of ECM.


Subject(s)
Immunologic Factors/administration & dosage , Malaria, Cerebral/prevention & control , Membrane Proteins/administration & dosage , Plasmodium berghei/immunology , Protozoan Proteins/administration & dosage , Recombinant Proteins/administration & dosage , Administration, Intravenous , Animals , Brain/pathology , Disease Models, Animal , Endothelial Cells/drug effects , Immunologic Factors/isolation & purification , Lymphocyte Activation , Malaria, Cerebral/pathology , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Protozoan Proteins/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , T-Lymphocytes, Regulatory/drug effects , Treatment Outcome
3.
Malar J ; 16(1): 458, 2017 Nov 13.
Article in English | MEDLINE | ID: mdl-29132428

ABSTRACT

BACKGROUND: A vaccine that targets multiple developmental stages of malaria parasites would be an effective tool for malaria control and elimination. METHODS: A conserved gene in Plasmodium, the Plasmodium berghei gene (PBANKA_020570) encoding a 51 kDa protein (pb51 gene), was identified through search of the PlasmoDB database using a combination of expression and protein localization criteria. A partial domain of the Pb51 protein was expressed in a prokaryotic expression system (rPb51) and used for immunization in mice. The protein expression profile and localization were studied by Western blot and indirect immunofluorescence assay (IFA), respectively. The inhibitory effect of the anti-rPb51 antibodies on parasite proliferation was evaluated in erythrocytes in vivo. The transmission-blocking activity of the immune sera was determined by in vitro ookinete conversion assay and by direct mosquito feeding assay (DFA). RESULTS: The rPb51 elicited specific antibodies in mice. Western blot confirmed Pb51 expression in schizonts, gametocytes and ookinetes. IFA showed localization of Pb51 on the outer membranes of schizonts, gametocytes, zygotes, retorts, ookinetes and sporozoites of P. berghei. Mice immunized with the rPb51 protein significantly reduced parasite proliferation and gametocyte conversion in vivo. Moreover, the rPb51 antisera also significantly reduced the in vitro ookinete conversion when added into the ookinete culture medium. In DFA, mice immunized with the rPb51 reduced the prevalence of mosquito infection by 21.3% and oocyst density by 54.8%. CONCLUSIONS: In P. berghei, P51 was expressed in both asexual erythrocytic and sexual stages and localized on the surface of these stages with the exception of the ring stage. The anti-rPb51 antibodies inhibited both P. berghei proliferation in mice and transmission of the parasite to mosquitoes.


Subject(s)
Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , Protozoan Proteins/genetics , Animals , Blotting, Western , Cell Proliferation/drug effects , Female , Fluorescent Antibody Technique, Indirect , Malaria/parasitology , Malaria/transmission , Mice , Mice, Inbred BALB C , Plasmodium berghei/genetics , Protozoan Proteins/metabolism , Rabbits
4.
Malar J ; 15(1): 537, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27821166

ABSTRACT

BACKGROUND: Artemisinin resistance in Plasmodium falciparum has emerged as a major threat for malaria control and elimination worldwide. Mutations in the Kelch propeller domain of PfK13 are the only known molecular markers for artemisinin resistance in this parasite. Over 100 non-synonymous mutations have been identified in PfK13 from various malaria endemic regions. This study aimed to investigate the genetic diversity of PvK12, the Plasmodium vivax ortholog of PfK13, in parasite populations from Southeast Asia, where artemisinin resistance in P. falciparum has emerged. METHODS: The PvK12 sequences in 120 P. vivax isolates collected from Thailand (22), Myanmar (32) and China (66) between 2004 and 2008 were obtained and 353 PvK12 sequences from worldwide populations were retrieved for further analysis. RESULTS: These PvK12 sequences revealed a very low level of genetic diversity (π = 0.00003) with only three single nucleotide polymorphisms (SNPs). Of these three SNPs, only G581R is nonsynonymous. The synonymous mutation S88S is present in 3% (1/32) of the Myanmar samples, while G704G and G581R are present in 1.5% (1/66) and 3% (2/66) of the samples from China, respectively. None of the mutations observed in the P. vivax samples were associated with artemisinin resistance in P. falciparum. Furthermore, analysis of 473 PvK12 sequences from twelve worldwide P. vivax populations confirmed the very limited polymorphism in this gene and detected only five distinct haplotypes. CONCLUSIONS: The PvK12 sequences from global P. vivax populations displayed very limited genetic diversity indicating low levels of baseline polymorphisms of PvK12 in these areas.


Subject(s)
Genetic Variation , Malaria, Vivax/parasitology , Plasmodium vivax/genetics , Protozoan Proteins/genetics , China , DNA, Protozoan/chemistry , DNA, Protozoan/genetics , Humans , Mutation , Myanmar , Plasmodium vivax/isolation & purification , Sequence Analysis, DNA , Thailand
5.
J Immunol ; 193(3): 1314-23, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24965778

ABSTRACT

In animal models of experimental cerebral malaria (ECM), neuropathology is associated with an overwhelming inflammatory response and sequestration of leukocytes and parasite-infected RBCs in the brain. In this study, we explored the effect of vitamin D (VD; cholecalciferol) treatment on host immunity and outcome of ECM in C57BL/6 mice during Plasmodium berghei ANKA (PbA) infection. We observed that oral administration of VD both before and after PbA infection completely protected mice from ECM. VD administration significantly dampened the inducible systemic inflammatory responses with reduced circulating cytokines IFN-γ and TNF and decreased expression of these cytokines by the spleen cells. Meanwhile, VD also resulted in decreased expression of the chemokines CXCL9 and CXCL10 and cytoadhesion molecules (ICAM-1, VCAM-1, and CD36) in the brain, leading to reduced accumulation of pathogenic T cells in the brain and ultimately substantial improvement of the blood-brain barriers of PbA-infected mice. In addition, VD inhibited the differentiation, activation, and maturation of splenic dendritic cells. Meanwhile, regulatory T cells and IL-10 expression levels were upregulated upon VD treatment. These data collectively demonstrated the suppressive function of VD on host inflammatory responses, which provides significant survival benefits in the murine ECM model.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Immunosuppressive Agents/administration & dosage , Malaria, Cerebral/immunology , Malaria, Cerebral/prevention & control , Plasmodium berghei/immunology , Vitamin D/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Dendritic Cells/immunology , Dendritic Cells/pathology , Down-Regulation/immunology , Female , Growth Inhibitors/administration & dosage , Growth Inhibitors/therapeutic use , Host-Parasite Interactions/immunology , Immunosuppressive Agents/therapeutic use , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Primary Cell Culture , Random Allocation , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/immunology , Vitamin D/therapeutic use
6.
Malar J ; 13: 116, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24670210

ABSTRACT

BACKGROUND: Malaria and schistosomiasis are endemic and co-exist in the same geographic areas, even co-infecting the same host. Previous studies have reported that concomitant infection with Schistosoma japonicum could offer protection against experimental cerebral malaria (ECM) in mice. This study was performed to evaluate whether alterations in parasite density could alter this protective effect. METHODS: Mice were inoculated with 100 or 200 S. japonicum cercariae followed by infection with high or low density of Plasmodium berghei ANKA strain eight weeks after the first infection. Then, parasitaemia, survival rate and blood-brain-barrier (BBB) damage were assessed. Interferon-gamma (IFN-γ), interleukin (IL)-4, IL-5, IL-13, IL-10, and TGF-ß levels were determined in splenocyte supernatants using enzyme-linked immunosorbent assay (ELISA). Cell surface/intracellular staining and flow cytometry were used to analyse the level of CD4(+)/CD8(+) T cells, CD4(+)CD25(+)Foxp3(+) Tregs, IL-10-secreting Tregs, and IL-10(+)Foxp3-CD4(+) T cells in the spleen, and CD4(+)/CD8(+) T cells infiltrating the brain. RESULTS: Co-infection with low density P. berghei and increased S. japonicum cercariae significantly increased the levels of IL-4, IL-5, IL-13, TGF-ß and Tregs, but significantly decreased the levels of IFN-γ and the percentage of CD4(+) T cells and CD8(+) T cells in the spleen and CD8(+) T cell infiltration in the brain. Increased worm loads also significantly decreased mortality and BBB impairment during ECM. When challenged with higher numbers of P. berghei and increased cercariae, the observed cytokine changes were not statistically significant. The corresponding ECM mortality and BBB impairment also remained unchanged. CONCLUSIONS: This study demonstrates that protection for ECM depends on the numbers of the parasites, S. japonicum and P. berghei, during co-infection. Alterations in the regulatory response appear to play a key role in this adaptation.


Subject(s)
Coinfection/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Schistosoma japonicum/immunology , Schistosomiasis japonica/immunology , Animals , Coinfection/parasitology , Coinfection/pathology , Cytokines/immunology , Disease Susceptibility/immunology , Disease Susceptibility/parasitology , Disease Susceptibility/pathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immune Tolerance , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Parasitemia/immunology , Parasitemia/parasitology , Parasitemia/pathology , Schistosomiasis japonica/parasitology , Schistosomiasis japonica/pathology , Spleen/immunology
7.
Cardiovasc Ultrasound ; 12: 21, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24950856

ABSTRACT

BACKGROUND: We previously reported the use of a catheter system to damage the tricuspid valve and create infectious endocarditis (IE) in an animal model. The current study aims to create a faint IE model suitable for antibiotic prophylaxis using a low bacterial inoculum. We also aim to explore a way to quantitatively assess valvular impairment and to predict the success of the IE models during catheterization. METHODS: Ninety rabbits were assigned to two groups according to the density of bacteria inoculated (1 × 10(5) CFU for Group A and 1 × 10(4) CFU for Group B). A catheter system consisting of a polyethylene catheter and a guide wire were used to damage the valve. The catheter system was passed through the rabbits' tricuspid valves under echocardiographic guidance. A pressure transducer was used to assess right atrial pressure (P(RA)) before and just after valvular damage to calculate the pressure alterations (ΔP(RA)). The animals in group A and B were divided into 3 subgroups according to the ΔP(RA) (0-5 mmHg for Groups A1 and B1; 5-10 mmHg for Groups A2 and B2; 10-15 mmHg for Groups A3 and B3). Staphylococcus aureus (ATCC 29213) inoculation was performed 24 hr after cardiac catheterization. RESULTS: Faint IE was confirmed in 20%, 93.3%, 26.7%, 6.7%, 20%, and 33.3% of the rabbits in Groups A1, A2, A3, B1, B2, and B3, respectively. There was no difference in the LV/RV ratio and VTR of the No-IE, faint-IE, and severe IE animals. Faint IE rabbits had a larger ΔPRA than No-IE rabbits (7.81 ± 1.21 vs. 2.48 ± 1.0, P < 0.01, for Group A; 7.60 ± 1.32 vs. 2.98 ± 1.08, P < 0.01, for Group B). The ΔPRA of severe IE and faint IE rabbits was significantly different (13.11 ± 1.31 vs. 7.81 ± 1.21, P < 0.01, for Group A; 12.73 ± 1.44 vs.7.60 ± 1.32, P < 0.01, for Group B). CONCLUSION: ΔP(RA) could be used to assess valvular impairment. Controlling the value of ΔP(RA) during catheterization and inoculating of an appropriate dose of bacteria was associated with a successful IE model.


Subject(s)
Atrial Pressure/physiology , Cardiac Catheterization/methods , Echocardiography/methods , Endocarditis, Bacterial/diagnostic imaging , Tricuspid Valve Insufficiency/diagnostic imaging , Animals , Atrial Function, Right/physiology , Disease Models, Animal , Endocarditis, Bacterial/microbiology , Female , Male , Predictive Value of Tests , Rabbits , Staphylococcal Infections/diagnostic imaging , Staphylococcus aureus , Tricuspid Valve/diagnostic imaging , Tricuspid Valve/microbiology
8.
Malar J ; 12: 322, 2013 Sep 14.
Article in English | MEDLINE | ID: mdl-24034228

ABSTRACT

BACKGROUND: Since helminths and malaria parasites are often co-endemic, it is important to clarify the immunoregulatory mechanism that occurs during the process of co-infection. A previous study confirmed that dendritic cells (DCs) are involved in the establishment and regulation of the T-cell-mediated immune response to malaria infection. In the current study, distinct response profiles for splenic DCs and regulatory T cell (Treg) responses were assessed to evaluate the effects of a pre-existing Schistosoma japonicum infection on malaria infection. METHODS: Malaria parasitaemia, survival rate, brain histopathology and clinical experimental cerebral malaria (ECM) were assessed in both Plasmodium berghei ANKA-mono-infected and S. japonicum-P. berghei ANKA-co-infected mice. Cell surface/intracellular staining and flow cytometry were used to analyse the level of splenic DC subpopulations, toll-like receptors (TLRs), DC surface molecules, Tregs (CD4⁺CD25⁺Foxp3⁺), IFN-γ/IL-10-secreting Tregs, and IFN-γ⁺/IL-10⁺-Foxp3⁻CD4⁺ T cells. IFN-γ, IL-4, IL-5, IL-10 and IL-13 levels were determined in splenocyte supernatants using enzyme-linked immunosorbent assay (ELISA). RESULTS: The co-infected mice had significantly higher malaria parasitaemia, compared with the mono-infected mice, on days 2, 3, 7 and 8 after P. berghei ANKA infection. Mono-infected mice had a slightly lower survival rate, while clinical ECM symptoms, and brain pathology, were significantly more severe during the period of susceptibility to ECM. On days 5 and 8 post P. berghei ANKA infection, co-infected mice had significantly lower levels of CD11c⁺CD11b⁺, CD11c⁺CD45R/B220⁺, CD11c⁺TLR4⁺, CD11c⁺TLR9⁺, CD11c⁺MHCII⁺, CD11c⁺CD86⁺, IFN-γ-secreting Tregs, and IFN-γ⁺Foxp3⁻CD4⁺ T cells in single-cell suspensions of splenocytes when compared with P. berghei ANKA-mono-infected mice. Co-infected mice also had significantly lower levels of IFN-γ and higher levels of IL-4, IL-5, and IL-13 in splenocyte supernatants compared to mono-infected mice. There were no differences in the levels of IL-10-secreting Tregs or IL-10⁺Foxp3⁻CD4⁺ T cells between co-infected and mono-infected mice. CONCLUSIONS: A Tregs-associated Th2 response plays an important role in protecting against ECM pathology. Pre-existing S. japonicum infection suppressed TLR ligand-induced DC maturation and had an anti-inflammatory effect during malaria infection not only by virtue of its ability to induce Th2 responses, but also by directly suppressing the ability of DC to produce pro-inflammatory mediators.


Subject(s)
Malaria/immunology , Plasmodium berghei/immunology , Schistosoma japonicum/immunology , Schistosomiasis japonica/immunology , Animals , Brain/pathology , Dendritic Cells/immunology , Disease Models, Animal , Female , Histocytochemistry , Immune Tolerance , Immunophenotyping , Malaria/parasitology , Malaria/pathology , Mice , Mice, Inbred C57BL , Parasitemia/parasitology , Schistosomiasis japonica/complications , Schistosomiasis japonica/pathology , Spleen/immunology , Survival Analysis , T-Lymphocytes, Regulatory/immunology
9.
Cardiovasc Ultrasound ; 11: 3, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23317050

ABSTRACT

BACKGROUND: The most widely used experimental models of infective endocarditis (IE) are the rabbit and rat models, in which cardiac valve lesions are induced by a polyethylene catheter introduced into the left ventricle through the aortic valve. Our study was designed to create a rabbit model of IE right-sided with echocardiographic guidance. METHODS: Thirty rabbits underwent both catheterization and inoculation (group A). These were divided into three subgroups of ten based on the time of catheter removal (immediately, after 24 h, and after death or moribundity for groups, A(1), A(2), and A(3), respectively). Ten inoculated-only and ten catheterized-only rabbits served as controls. A catheter system consisted of a polyethylene catheter and a guide wire inside it. This system was passed through the rabbits' tricuspid valves under echocardiographic guidance to damage them. The ratio of left ventricle to right ventricle (LV/RV) was measured in a four-chamber view before catheterization and at the time of death or moribundity. The peak velocity of tricuspid regurgitation (V(TR)) was measured in a four-chamber view at the time of catheterization and at the time of death or moribundity. Staphylococcus aureus (ATCC 29213) inoculation was performed 24 h after right heart catheterization to produce IE. RESULTS: IE was confirmed in 28 of 30 rabbits by macroscopic and histologic examination of tricuspid valves, blood cultures, and bacterial count in cardiac vegetations. Cardiac vegetations were confirmed in 25 of 28 IE rabbits by echocardiography. Enlargement of right ventricle dimension with a significantly decreased LV/RV ratio was confirmed in all IE rabbits at the time of death or moribundity than at the initial state (1.11 ± 0.35 vs. 1.95 ± 0.39, P < 0.01; 1.21 ± 0.34 vs. 1.98 ± 0.35, P < 0.01; 1.04 ± 0.31 vs. 2.00 ± 0.41, P < 0.01 for groups A(1), A(2), and A(3), respectively). V(TR) was significantly higher in all the IE rabbits at the time of death or moribundity than at the time of catheterization (1.89 ± 0.46 vs 0.76 ± 0.45, P < 0.01; 2.04 ± 0.73 vs 0.68 ± 0.66, P < 0.01; 2.24 ± 0.51 vs 0.87 ± 0.55, P < 0.01 for group A(1), A(2), and A(3), respectively). CONCLUSION: The models described herein closely reproduced the pathogenesis and pathophysiology of right heart catheter-induced endocarditis in humans. Echocardiographic guidance is helpful in the process of right heart catheterization. Some echocardiographic parameters, such as V(TR) and the LV/RV ratio could be used to assess the success or failure of the IE models.


Subject(s)
Echocardiography/methods , Endocarditis, Bacterial/diagnostic imaging , Staphylococcal Infections/diagnostic imaging , Staphylococcus aureus/isolation & purification , Animals , Cardiac Catheterization/adverse effects , Catheters/microbiology , Disease Models, Animal , Endocarditis, Bacterial/etiology , Endocarditis, Bacterial/microbiology , Female , Male , Rabbits , Staphylococcal Infections/etiology , Staphylococcal Infections/microbiology
10.
J Basic Microbiol ; 52(3): 332-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22052457

ABSTRACT

RNA interference (RNAi)-based short interfering RNAs (siRNAs) targeting the viral genes and the host cellular genes have been used to suppress Measles virus (MV) replication in vitro. In order to select suitable target genes and highly effective target sites for developing effective RNAi-mediated anti-MV therapy, in this study, nine short hairpin RNA (shRNA) expression vectors, which expressed siRNAs targeting the host celluar Rab9 GTPase gene, the viral large protein (L) gene and nucleoprotein (N) gene, respectively, were constructed and used to compare their ability to inhibit MV replication in Vero-E6 cells. The results showed that nine siRNAs targeting different genes exhibited different inhibitory efficacy on MV replication in vitro (about 23-94%), which could last at least 168 h post-infection. Of the nine siRNAs, R2, L1 and N2 more effectively decreased MV replication by over 90%. Furthermore, inhibitory efficacy on MV replication were increased and reached almost 100% when cells were transfected with pR2, pL1 and pN2 together. These results emphasis the importance of selecting suitable siRNA target sites for developing siRNAs-based drug therapy for MV, and demonstrate the potential of combination of siRNAs targeting different genes as a therapeutic approach to treat MV infection.


Subject(s)
Gene Silencing , Measles virus/growth & development , Measles virus/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Virus Replication , Animals , Antiviral Agents/metabolism , Chlorocebus aethiops , Genes, Viral , Measles virus/physiology , Time Factors , Vero Cells
11.
World J Microbiol Biotechnol ; 28(1): 381-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22806814

ABSTRACT

Amplifying the variable (Fv or V) regions of immunoglobulins (Ig) has become a challenge in cloning antibody genes for phage display, a technique used to study protein-protein, protein-peptide, and protein-DNA interactions using bacteriophages to connect proteins with the genetic information that encodes them. Key parameters affecting the amplification of full antibody repertoires includes the availability of primers that can amplify as many V genes as possible; however the strategy used to design these primers and programs used to make the necessary alignments have not been well studied and clearly detailed in the literature. Here, we present a set of primers computationally designed by iCODEHOP based on a database of human germline Ig sequences. We used reverse transcription polymerase chain reaction (RT-PCR) protocols that would recognize the V(H) genes from human peripheral blood mononuclear cells. We identified the most highly conserved region in framework 1 and framework 4 of the Ig cDNA, and designed a set of degenerated 5' primers. The V(H) genes were successfully amplified by RT-PCR. This new primer has facilitated the creation of more diverse V(H) libraries than has been previously possible. Moreover, iCODEHOP improved the primer design efficiency and was found useful both for cloning unknown genes in gene families and for building V(H) gene libraries.


Subject(s)
DNA Primers/genetics , Genes, Immunoglobulin Heavy Chain , Immunoglobulin Variable Region/genetics , Base Sequence , Cloning, Molecular , Computational Biology , Databases, Genetic , Humans , Reverse Transcriptase Polymerase Chain Reaction , Software
12.
Article in English | MEDLINE | ID: mdl-35792443

ABSTRACT

BACKGROUND: With the emergence of resistance to front-line antimalarials, there is an urgent need to develop new medicines, including those targeting sexual development. This study aimed to assess the activity of a panel of phosphatase inhibitors against the sexual development of Plasmodium berghei and evaluate their potential as transmission-blocking agents. METHODS: Twenty-five compounds were screened for transmission-blocking activity in vitro using the P. berghei ookinete culture assay. The inhibitory effects on male gametogenesis, gamete-ookinete, and zygote-ookinete formation were evaluated. The transmission-blocking activity of two compounds was evaluated using an in vivo mosquito feeding assay. Their cytotoxic effects were assessed on the human cell line HepG2. RESULTS: Twelve compounds inhibited P. berghei ookinete formation with an IC50 < 10 µM. Two compounds, BVT-948 and alexidine dihydrochloride, significantly inhibited different developmental stages from gametogenesis through ookinete maturation. They also showed a substantial in vivo transmission-blocking activity by the mosquito feeding assay. CONCLUSIONS: Some phosphatase inhibitors effectively inhibited Plasmodium sexual development and exhibited evident transmission-blocking activity, suggesting that phosphatases are valid targets for antimalarial development.


Subject(s)
Antimalarials , Culicidae , Malaria , Parasites , Animals , Antimalarials/pharmacology , Biguanides , Enzyme Inhibitors/pharmacology , Humans , Malaria/drug therapy , Male , Phosphoric Monoester Hydrolases/pharmacology , Plasmodium berghei , Sexual Development
13.
Int Immunopharmacol ; 90: 107052, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33310296

ABSTRACT

Numerous studies have focused on the treatment of melanoma, but the current therapies for melanoma have limited therapeutic effects. To find a more effective therapy for melanoma, we combined artificially designed CpG ODN (cytosine-phosphate-guanine oligodeoxynucleotides) and siRNAs (small-interfering ribonucleic acids) targeting PD-1 (programmed cell death protein 1), which were delivered by attenuated Salmonella to treat melanoma in mice, and explored the underlying antitumor mechanisms. We found that mice receiving the combination therapy had the smallest tumor size and the longest survival time. The possible mechanisms underlying this phenomenon include pathways mediated by cyclin D1, p-STAT3 (phosphorylated signal transducers and activators of transcription protein 3), MMP2 (matrix metallopeptidase 2) and cleaved caspase 3, since after treatment, the expression of cyclin D1, p-STAT3, and MMP2 decreased but that of cleaved caspase 3 increased; additional mechanisms include increases in the recruitment of immune cells to tumor sites and in the number of immune cells in mouse spleens and the upregulation of TNF-α (tumor necrosis factor) and IL-6 (interleukin 6). We demonstrated that the combination therapy composed of CpG ODN and PD-1-siRNA delivered by attenuated Salmonella exhibited a strong ability to inhibit melanoma and improve the antitumor immune responses of tumor-bearing mice.


Subject(s)
Antineoplastic Agents/pharmacology , Genetic Vectors , Melanoma, Experimental/therapy , Oligodeoxyribonucleotides/pharmacology , Programmed Cell Death 1 Receptor/genetics , RNA, Small Interfering/genetics , RNAi Therapeutics , Salmonella/genetics , Toll-Like Receptor 9/agonists , Animals , Cell Line, Tumor , Combined Modality Therapy , Cytokines/blood , Female , Gene Expression Regulation, Neoplastic , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/metabolism , Time Factors , Toll-Like Receptor 9/metabolism , Tumor Burden
14.
Parasit Vectors ; 14(1): 407, 2021 Aug 16.
Article in English | MEDLINE | ID: mdl-34399829

ABSTRACT

BACKGROUND: Plasmodium vivax transmission-blocking vaccines (TBVs) are receiving increasing attention. Based on excellent transmission-blocking activities of the PbPH (PBANKA_0417200) and PbSOP26 (PBANKA_1457700) antigens in Plasmodium berghei, their orthologs in P. vivax, PVX_098655 (PvPH) and PVX_101120 (PvSOP26), were selected for the evaluation of their potential as TBVs. METHODS: Fragments of PvPH (amino acids 22-304) and PvSOP26 (amino acids 30-272) were expressed in the yeast expression system. The recombinant proteins were used to immunize mice to obtain antisera. The transmission-reducing activities of these antisera were evaluated using the direct membrane feeding assay (DMFA) using Anopheles dirus mosquitoes and P. vivax clinical isolates. RESULTS: The recombinant proteins PvPH and PvSOP26 induced robust antibody responses in mice. The DMFA showed that the anti-PvSOP26 sera significantly reduced oocyst densities by 92.0 and 84.1% in two parasite isolates, respectively, whereas the anti-PvPH sera did not show evident transmission-reducing activity. The variation in the DMFA results was unlikely due to the genetic polymorphisms of the two genes since their respective sequences were identical in the clinical P. vivax isolates. CONCLUSION: PvSOP26 could be a promising TBV candidate for P. vivax, which warrants further evaluation.


Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Animals , Female , Humans , Immunogenicity, Vaccine , Malaria Vaccines/genetics , Malaria, Vivax/parasitology , Malaria, Vivax/transmission , Mice , Mice, Inbred BALB C , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccination/methods , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Yeasts/genetics
15.
Can J Microbiol ; 56(1): 77-80, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20130697

ABSTRACT

Rab9 has been identified as a key component for the replication of measles virus (MV). In this study, gene-specific shRNAs were developed to suppress the replication of MV in culture cells by silencing the expression of Rab9 GTPase gene. Rab9 GTPase gene-specific shRNAs were designed and cloned into the expression vector of pSUPER.neo+EGFP. Vero-E6 cells were transfected with the recombinant plasmid via liposome and then infected with MV. The expression of Rab9 GTPase mRNA and protein were assayed by RT-PCR and Western blotting, respectively. ShRNA-mediated inhibition of MV replication was further evaluated by detecting the titer of MV. The results showed that the expression of Rab9 GTPase was dramatically and stably downregulated by the generated shRNAs targeting Rab9 GTPase gene, which contribute to the inhibition of MV replication, indicating these shRNAs could be potentially developed into therapeutic agents for the treatment of MV infection in the future.


Subject(s)
Gene Expression Regulation, Viral/drug effects , Measles virus/physiology , RNA, Small Interfering/pharmacology , Virus Replication/drug effects , Animals , Chlorocebus aethiops , Down-Regulation , Measles/drug therapy , Measles/virology , Measles virus/genetics , Measles virus/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Vero Cells , Virus Replication/genetics
16.
Acta Med Okayama ; 64(1): 49-54, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20200584

ABSTRACT

In Liaoning Province in northeastern China, we found a G6PD-deficient patient at the age of 3. By the classification of the World Health Organization, this patient was categorized as class I (very severe G6PD deficiency). When we investigated the G6PD gene of the patient, we found that he had a replacement of G to A at nucleotide 1339. As a result, the amino acid at position 447 should change from Gly to Arg. This replacement is known as G6PD Santiago de Cuba, because it was first discovered in a Cuban boy who showed heavy chronic anemia. Today, 28 G6PD variants have been reported in the Chinese population, and all are categorized as class II (severe deficiency) or class III (mild deficiency); in class II or III deficiency, anemia is not present in daily life, but hemolytic attack can occur when the carrier ingests certain oxidative medicines or foods. This is the first report of a G6PD-deficient Chinese patient in the category of class I. We intended to find other G6PD-deficient cases in northeastern China and tested several hundred blood samples, but no cases of G6PD deficiency were found (0/414). In central China, where falciparum malaria was endemic from the 1950s to 1970s, we found two G6PD-deficient cases (2/27) and the other members from their families whose variant type was G6PD Kaiping (1388G > T), which is a common variant in the Chinese population.


Subject(s)
Asian People/genetics , Asian People/statistics & numerical data , Glycogen Storage Disease Type I/ethnology , Glycogen Storage Disease Type I/genetics , Anemia, Hemolytic/ethnology , Anemia, Hemolytic/genetics , Child, Preschool , China/epidemiology , Family Health , Health Surveys , Humans , Male , Pedigree , Point Mutation , Severity of Illness Index
17.
Peptides ; 133: 170375, 2020 11.
Article in English | MEDLINE | ID: mdl-32771373

ABSTRACT

Lipotoxicity has been implicated in many disease processes, and prolonged exposure to high lipid levels often leads to the activation of a variety of abnormal signals, which in turn leads to the induction of inflammation. The aim of our study was to explore the correlation between mammalian target of rapamycin (mTOR) and inflammation by studying high-fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) in rats and palmitate (PA)-induced inflammation (lipotoxicity) in HepG2 cells. In addition, we investigated whether the glucagon-like peptide-1 (GLP-1) analogue liraglutide can protect rats and HepG2 cells from lipotoxicity. Our results showed that an HFD and PA significantly increased inflammation by activating the mTORC1 pathway in vitro and in vivo. Treatment with rapamycin (an mTOR inhibitor) inhibited some effects of PA on inflammation. Furthermore, we observed that liraglutide inhibited PA-induced inflammation by inactivating mTORC1 signalling molecules. Overall, our findings demonstrated that mTORC1 signalling pathways were involved primarily in high lipid level-induced inflammation. Importantly, liraglutide may protect against lipotoxicity-induced inflammation by regulating mTORC1-dependent pathways.


Subject(s)
Hepatitis/drug therapy , Liraglutide/pharmacology , Mechanistic Target of Rapamycin Complex 1/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Body Weight/drug effects , Diet, High-Fat/adverse effects , Eating/drug effects , Hep G2 Cells , Hepatitis/etiology , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Insulin Resistance , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Palmitates/toxicity , Rats, Sprague-Dawley
18.
Appl Immunohistochem Mol Morphol ; 28(8): 602-610, 2020 09.
Article in English | MEDLINE | ID: mdl-31394555

ABSTRACT

The p21-activated kinase (PAK) family of serine/threonine kinases plays a pivotal role in various human tumors, as supported by our previous report on the overexpressed PAK isoforms in non-small cell lung cancer (NSCLC). To better understand the role of PAKs in tumorigenesis, the authors examined PAK1 expression patterns and its significance in NSCLC. It was demonstrated by immunohistochemical staining that PAK1 was increased and localized in the cytoplasm in 151 of 207 cases. High levels of PAK1 expression correlated with a histologic type of tumor (squamous cell carcinoma), tumor node metastasis stage, and lymph nodal status. We also examined the biological role of PAK1 in lung cancer cell lines transfected with PAK1-small interfering RNA. Decreased expression of PAK1 inhibited lung cancer cell proliferation and invasion, which is the major cause of lung cancer malignancy. Downregulated expression of PAK1 hampered rapidly accelerated fibrosarcoma/mitogen-activated extracellular signal-regulated kinase/extracellular signal-regulated kinase pathway activity but did not affect Wnt/ß-catenin signaling. Our findings suggest that PAK1 is an important oncogene in NSCLC, as decreased expression of PAK1 inhibited the proliferation and invasion of NSCLC cells by blocking the ERK pathway. These results provide evidence for using PAK1 inhibition as potential anticancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Proliferation/genetics , Lung Neoplasms/metabolism , MAP Kinase Signaling System/genetics , p21-Activated Kinases/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cytoplasm/metabolism , Down-Regulation , Female , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , RNA, Small Interfering , Wnt Signaling Pathway/genetics , p21-Activated Kinases/genetics
19.
Article in English | MEDLINE | ID: mdl-32670896

ABSTRACT

Quiescin sulfhydryl oxidase (QSOX), present in a wide variety of eukaryotic species, catalyzes the insertion of disulfide bonds into unfolded, reduced proteins. Here we characterized the QSOX protein from the rodent malaria parasite Plasmodium berghei (PbQSOX), which is conserved in all sequenced malaria parasite species. The PbQSOX protein was not expressed in asexual erythrocytic stages, but was most abundantly expressed in ookinetes. Indirect immunofluorescence assays revealed PbQSOX was not only localized in cytoplasm of gametocytes, gametes and ookinetes, but also expressed on the surface of gametes and ookinetes. Western blot identified extracellular presence of PbQSOX in the culture medium of ookinetes suggestive of secretion. Pbqsox deletion (Δpbqsox) did not affect asexual intraerythrocytic development, but reduced exflagellation of male gametocytes as well as formation and maturation of ookinetes. Pbqsox deletion also led to a significant increase in the reduced thiol groups of ookinete surface proteins, suggesting that it may play a role in maintaining the integrity of disulfide bonds of surface proteins, which might be needed for ookinete development. Mosquitoes that fed on Δpbqsox-infected mice showed a significant reduction in ookinete and oocyst numbers compared to those fed on wild-type parasite-infected mice. Further, both polyclonal mouse antisera and a monoclonal antibody against the recombinant PbQSOX exhibited substantial transmission-blocking activities in in vitro and mosquito feeding assays, suggesting QSOX is a potential target for blocking parasite transmission.


Subject(s)
Malaria , Parasites , Animals , Male , Mice , Oocysts , Oxidoreductases , Plasmodium berghei
20.
Int Immunopharmacol ; 74: 105683, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31220697

ABSTRACT

The antiviral drug Telbivudine (LdT) has an extrahepatic pharmaceutical effect that improves renal inflammation and tubulointerstitial fibrosis. However, the exact mechanism of action requires further investigation. Toll-like receptor 4 (TLR4) is involved in several physiological processes, including inflammation, fibrosis, innate immunity, and hepatitis B virus-associated glomerulonephritis. The epithelial-to-mesenchymal transition (EMT) is the characteristic pathological change in tubulointerstitial fibrosis. In this study, we used transforming growth factor-ß (TGF-ß) to stimulate human proximal tubular epithelial (HK-2) cells to investigate the effects of LdT in EMT. In addition, we treated HK-2 cells with a TLR4 agonist, lipopolysaccharide, to determine the effect of LdT on TLR4. The results indicated that LdT inhibited the expression of TLR4 and its downstream proteins. It also decreased the release of inflammatory factors, downregulated the TGF-ß/Smad signaling pathway, and reversed the EMT changes seen in HK-2 cells. In conclusion, LdT antagonized TLR4 to inhibit EMT in proximal tubular epithelial cells.


Subject(s)
Antiviral Agents/pharmacology , Epithelial Cells/physiology , Kidney Tubules, Proximal/pathology , Renal Insufficiency, Chronic/drug therapy , Telbivudine/pharmacology , Cell Line , Epithelial-Mesenchymal Transition , Fibrosis , Humans , Immunity, Innate , Lipopolysaccharides/immunology , Signal Transduction , Smad Proteins/metabolism , Toll-Like Receptor 4/metabolism , Transforming Growth Factor beta/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL