Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Clin Immunol ; 257: 109843, 2023 12.
Article in English | MEDLINE | ID: mdl-37981106

ABSTRACT

Methicillin-resistant Staphylococcus aureus, poses a significant threat through infections in both community and hospital settings. To address this challenge, we conducted a phase I clinical trial study involving a recombinant Staphylococcus aureus vaccine. Utilizing peripheral blood lymphocytes from 64 subjects, we isolated antigen-specific memory B cells for subsequent single-cell sequencing. Among the 676 identified antigen-binding IgG1+ clones, we selected the top 10 antibody strains for construction within expression vectors. Successful expression and purification of these monoclonal antibodies led to the discovery of a highly expressed human antibody, designated as IgG-6. This antibody specifically targets the pentameric form of the Staphylococcus aureus protein A (SpA5). In vivo assessments revealed that IgG-6 provided prophylactic protection against MRSA252 infection. This study underscores the potential of human antibodies as an innovative strategy against Staphylococcus aureus infections, offering a promising avenue for further research and clinical development.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Staphylococcus aureus , Humans , Antibodies, Bacterial , Antibodies, Monoclonal , Immunoglobulin G , Methicillin-Resistant Staphylococcus aureus/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
2.
Biomaterials ; 292: 121907, 2023 01.
Article in English | MEDLINE | ID: mdl-36436305

ABSTRACT

The ongoing SARS-CoV-2 pandemic represents a brutal reminder of the continual threat of mucosal infectious diseases. Mucosal immunity may provide robust protection at the predominant sites of SARS-CoV-2 infection. However, it remains unclear whether respiratory mucosal administration of DNA vaccines could confer protective immune responses against SARS-CoV-2 challenge due to insurmountable barriers posed by the airway. Here, we applied self-assembled peptide-poloxamine nanoparticles with mucus-penetrating properties for pulmonary inoculation of a COVID-19 DNA vaccine (pSpike/PP-sNp). The pSpike/PP-sNp not only displays superior gene transfection and favorable biocompatibility in the mouse airway, but also promotes a tripartite immunity consisting of systemic, cellular, and mucosal immune responses that are characterized by mucosal IgA secretion, high levels of neutralizing antibodies, and resident memory phenotype T-cell responses in the lungs of mice. Most importantly, immunization with pSpike/PP-sNp completely eliminates SARS-CoV-2 infection in both upper and lower respiratory tracts and enables 100% survival rate of mice following lethal SARS-CoV-2 challenge. Our findings indicate PP-sNp is a promising platform in mediating DNA vaccines to elicit all-around mucosal immunity against SARS-CoV-2.


Subject(s)
COVID-19 , Nanoparticles , Vaccines, DNA , Mice , Animals , SARS-CoV-2 , Antibodies, Viral , COVID-19/prevention & control , Vaccination , Peptides , DNA , Antibodies, Neutralizing
3.
J Immunol Res ; 2022: 1403788, 2022.
Article in English | MEDLINE | ID: mdl-36471700

ABSTRACT

Tissue-resident memory T (TRM) cells are immune sentinels that bear a key role in the local immune system and rapidly respond to infection. Our previous studies showed that mucosal immunization via intranasal pathways was more effective than intramuscular route. However, the mechanism of enhanced protective immunity remains unclear. Here, we formulated a Pseudomonas aeruginosa vaccine composed of type III secretion protein PcrV from P. aeruginosa and curdlan adjuvant and then administered by the intranasal route. Flow cytometry and immunofluorescence staining showed that the ratio of CD44+CD62L-CD69+CD4+ TRM cells induced by this vaccine was significantly increased, and IL-17A production was notably enhanced. Further analysis revealed that vaccinated mice can protect against the P. aeruginosa challenge even after administration with FTY720 treatment. What is more, our results showed that CD4+ TRM might be involved in the recruitment of neutrophils and provided partial protection against Pseudomonas aeruginosa. Taken together, these data demonstrated that CD4+ TRM cells were elicited in lung tissues after immunization with rePcrV and contributed to protective immunity. Furthermore, it provided novel strategies for the development of vaccines for P. aeruginosa and other respiratory-targeted vaccines.


Subject(s)
Pseudomonas aeruginosa , Vaccines , Mice , Animals , Immunologic Memory , Memory T Cells , Vaccination , Lung , Administration, Intranasal
4.
Oncogenesis ; 11(1): 29, 2022 May 31.
Article in English | MEDLINE | ID: mdl-35641480

ABSTRACT

The latest study shows that gastric cancer (GC) ranked the fifth most common cancer (5.6%) with over 1 million estimated new cases annually and the fourth most common cause of cancer death (7.7%) globally in 2020. Metastasis is the leading cause of GC treatment failure. Therefore, clarifying the regulatory mechanisms for GC metastatic process is necessary. In the current study, we discovered that calreticulin (CALR) was highly expressed in GC tissues and related to lymph node metastasis and patient's terrible prognosis. The introduction of CALR dramatically promoted GC cell migration in vitro and in vivo, while the repression of CALR got the opposite effects. Cell migration is a functional consequence of the epithelial-mesenchymal transition (EMT) and is related to adhesion of cells. Additionally, we observed that CALR inhibition or overexpression regulated the expression of EMT markers (E-cadherin, ZO-1, Snail, N-cadherin, and ZEB1) and cellular adhesive moleculars (Fibronectin, integrin ß1and MMP2). Mechanistically, our data indicated that CALR could mediate DNA methylation of E-cadherin promoter by interacting with G9a, a major euchromatin methyltransferase responsible for methylation of histone H3 on lysine 9(H3K9me2) and recruiting G9a to the E-cadherin promoter. Knockdown of G9a in CALR overexpressing models restored E-cadherin expression and blocked the stimulatory effects of CALR on GC cell migration. Taken together, these findings not only reveal critical roles of CALR medicated GC metastasis but also provide novel treatment strategies for GC.

5.
J Cell Physiol ; 236(1): 536-548, 2021 01.
Article in English | MEDLINE | ID: mdl-32557622

ABSTRACT

Although the incidence and mortality of gastric cancer (GC) are slowly decreasing, the overall prognosis of GC patients with distal metastasis remains dismal. Long non-coding RNA PVT1 has been verified to function as a tumor promoter in several types of cancer. However, the role of PVT1 in GC metastasis remains obscure. Herein, we found that PVT1 was highly expressed in GC tissues and high PVT1 level was associated with tumor stage, lymph node metastasis, and poor prognosis. Overexpression of PVT1 significantly elevated epithelial-to-mesenchymal transition (EMT) marker (N-cadherin, ZEB1, and ZEB2) levels and promoted GC cell EMT process and tumor metastasis in vitro and in vivo. Mechanistically, Snail was identified as a direct target of miR-30a. PVT1 could bind with miR-30a and increase the expression of Snail by acting as a competing endogenous RNA, whereas re-expression of miR-30a in GC cells rescued the EMT markers, decreased Snail level, and inhibited GC cell migration. Taken together, these findings provide a new light on PVT1 in the pathogenesis and development of GC and an important implication for future therapy of the GC.


Subject(s)
Cell Movement/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Biomarkers, Tumor/genetics , Cadherins/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , MicroRNAs/genetics , Prognosis , Stomach Neoplasms/pathology
6.
Microb Pathog ; 152: 104650, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33232766

ABSTRACT

To provide detail data for Campylobacter jejuni (C.jejuni) vaccine research, this study performed epitope prediction analysis technology to screen the B cell immunodominant epitopes of C. jejuni adhesion protein PEB1 and evaluated the immunoprotective effect. The overlapping peptides were synthesized and B-cell immunodominant epitopes of PEB1 were identified by ELISA. BALB/c mice were immunized with the immunodominant epitopes of PEB1 conjugated with KLH plus CFA/IFA. The titers for immunodominant peptide antiserum against PEB1 were detected by ELISA. The bacterial colonization and the relative expression level of TNF-α were analyzed after the mice challenged with C. jejuni 11,168. The function of antibody induced by immunodominant PEB1 epitopes were performed by opsonophagocytic killing. The results showed that PEB155-72aa, PEB197-114aa, PEB1211-228aa were the immunodominant peptides and could induce strong B cell mediated humoral immunity response. Antiserum from the immunodominant peptides group significantly enhanced opsonize phagocytosis than CFA/IFA group (P<0.01). Both the bacterial burdens and the TNF-α expression level in the immunodominant peptides groups were significantly lower than those in the control group (P<0.01). Moreover, the immune protective effect of the three immunodominant peptides depended on the B cell immunity response in vivo study. In conclusion, three specific B cell immunodominant epitopes with good immunogenicity and immunoprotection efficacy were successfully identified, indicating that could be used in the anti- C. jejuni vaccine research and development.


Subject(s)
Campylobacter jejuni , Animals , B-Lymphocytes , Epitopes, B-Lymphocyte , Immunodominant Epitopes , Mice , Mice, Inbred BALB C
7.
Clin Biochem ; 84: 79-86, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32673627

ABSTRACT

BACKGROUND: Although stable microRNAs (miRNAs) are present in human peripheral blood and have been considered as novel biomarkers for various diseases. But there is little research about miRNAs as biomarkers of mesangial proliferative glomerulonephritis (MsPGN). This study aimed to identify whether there exist disordered circulating miRNAs that can function as biomarkers for MsPGN disease activity. METHODS: The candidate miRNAs were validated in 70 MsPGN patients and 70 healthy controls by quantitative real-time PCR (RT-qPCR). The specificity and sensitivity of the miRNA panel was assessed by receiver operating characteristic (ROC) curves. In addition, the candidate miRNA levels were measured in the different MsPGN progression and in the membranous nephropathy (MN) patients and the hypothetical role of the candidate miRNA on mesangial cell proliferation was analysed. Situ hybridization was performed to examine the candidate miRNA levels in the glomerulus. RESULTS: These results showed that miR-106a-5p and miR-30a-5p were highly expressed in MsPGN patients compared with healthy controls and could discriminate MsPGN from healthy controls with an area under the ROC curve (AUC) of 0.93. In addition, the two miRNAs were not only higher in moderate and severe MsPGN patients, but could distinguish MsPGN from MN. We also observed a decreased expression in MsPGN regression group after treatment. Plasma miR-106a-5p level was positively correlated with estimated glomerular filtration rate (eGFR). Furthermore, the two miRNAs were highly expressed in MsPGN glomerulus and their overexpression could prompt mesangial cell proliferation. CONCLUSION: Plasma miR-30a-5p and miR-106a-5p can serve as novel and potential diagnostic biomarkers for MsPGN.


Subject(s)
Glomerulonephritis, Membranoproliferative/blood , MicroRNAs/blood , Adult , Biomarkers, Tumor/blood , Circulating MicroRNA/blood , Circulating MicroRNA/genetics , Female , Gene Expression Profiling/methods , Glomerulonephritis/blood , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/pathology , Humans , Male , Mesangial Cells/metabolism , Mesangial Cells/pathology , MicroRNAs/genetics , Middle Aged , ROC Curve , Real-Time Polymerase Chain Reaction , Transcriptome
8.
Front Immunol ; 11: 806, 2020.
Article in English | MEDLINE | ID: mdl-32477338

ABSTRACT

During tumorigenesis, tumor infiltrating regulatory T (Treg) cells restrict the function of effector T cells in tumor microenvironment and thereby promoting tumor growth. The anti-tumor activity of effector T cells can be therapeutically unleashed, and is now being exploited for the treatment of various types of human cancers. However, the immune suppressive function of Treg cells remains a major hurdle to broader effectiveness of tumor immunotherapy. In this article, we reported that the deletion of Bcl6 specifically in Treg cells led to stunted tumor growth, which was caused by impaired Treg cell responses. Notably, Bcl6 is essential in maintaining the lineage stability of Treg cells in tumor microenvironment. Meanwhile, we found that the absence of follicular regulatory T (Tfr) cells, which is a result of Bcl6 deletion in Foxp3+ cells, was dispensable for tumor control. Importantly, the increased Bcl6 expression in Treg cells is associated with poor prognosis of human colorectal cancer and lymph node metastasis of skin melanoma. Furthermore, Bcl6 deletion in Treg cells exhibits synergistic effects with immune checkpoint blockade therapy. Collectively, these results indicate that Bcl6 actively participates in regulating Treg cell immune responses during tumorigenesis and can be exploited as a therapeutic target of anti-tumor immunity.


Subject(s)
Carcinogenesis/immunology , Colorectal Neoplasms/genetics , Immunity , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Skin Neoplasms/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Female , Gene Expression , Gene Knockout Techniques , Humans , Male , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Prognosis , Proto-Oncogene Proteins c-bcl-6/deficiency , Skin Neoplasms/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
9.
Cancer Biomark ; 27(4): 471-484, 2020.
Article in English | MEDLINE | ID: mdl-32116236

ABSTRACT

BACKGROUND: Gastric cancer is one of the leading causes of death worldwide. MicroRNA-30a (miR-30a) has been demonstrated to be involved in several types of cancer development. OBJECTIVE: We aimed to identify the molecular mechanism of miR-30a in gastric cancer. METHODS: We investigated the expression of miR-30a in gastric cancer tissues by qRT-PCR. The role of miR-30a on the metastasis and proliferation of gastric cancer was evaluated by cell migration assay, CCK-8 assay and tumor peritoneal dissemination model. The target of miR-30a in gastric cancer was identified. RESULTS: We discovered that miR-30a was significantly downregulated in gastric cancer tissues compared with adjacent nonmalignant tissues. The expression of miR-30a was inversely correlated with progression of gastric cancer. Gain- and loss-of function revealed that miR-30a acted as a potent tumor suppressor in gastric cancer. Re-expressed miR-30a inhibited gastric cancer cells migration, knock down miR-30a have the opposite effects. Furthermore, overexpression of miR-30a suppressed tumor peritoneal dissemination in vivo. We identified that fibroblast activation protein α (FAPα) was a direct target of miR-30a. The relative expression of FAPα was significantly higher in gastric cancer tissues compared with adjacent nonmalignant tissues. Inhibition of FAPα could recapitulate the effects of miR-30a, and overexpression of FAPα could abrogate the effect of miR-30a. CONCLUSION: MiR-30a inhibited gastric cancer metastasis by targeting FAPα, suggesting that miR-30a may function as a novel tumor suppressor in gastric cancer.


Subject(s)
Gelatinases/metabolism , Membrane Proteins/metabolism , MicroRNAs/genetics , Serine Endopeptidases/metabolism , Stomach Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Endopeptidases , Epithelial-Mesenchymal Transition , Female , Gelatinases/genetics , Genes, Tumor Suppressor , Humans , Lymphatic Metastasis , Male , Membrane Proteins/genetics , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Serine Endopeptidases/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Xenograft Model Antitumor Assays
10.
Oncogene ; 38(17): 3134-3150, 2019 04.
Article in English | MEDLINE | ID: mdl-30626935

ABSTRACT

Gastric cancer (GC) is the second leading cause of death among patients with cancer in China. The primary reason of GC treatment failure is metastasis. Therefore, identifying metastatic biomarkers and clarifying the regulatory mechanisms involved in the GC metastatic process are important. Here, we found that KIAA1199, a cell migration-inducing protein, was significantly overexpressed in GC and correlated with lymph node metastasis and poorer patient survival. Additionally, the introduction of KIAA1199 dramatically promoted GC cell proliferation and migration in vitro and in vivo, and the inhibition of KIAA1199 suppressed GC cell growth and migration and induced GC cell apoptosis. Cell migration is a functional consequence of the epithelial-mesenchymal transition (EMT). In this study, we found that KIAA1199 inhibition or overexpression regulated the expression of E-cadherin and N-cadherin through KIAA1199 binding to WW domain binding protein 11 (WBP11) and protein tyrosine phosphatase type IVA, member 3 (PTP4A3) and through the subsequent activation of the FGFR4/Wnt/ß-catenin and EGFR signaling pathways. More importantly, ectopic expression of WBP11 or PTP4A3 blocked the stimulatory effects of KIAA1199 on GC cell proliferation and migration. Meanwhile, we illustrated that KIAA1199 was a target gene of miR-29c-3p and that miR-29c-3p overexpression led to decreased migration of GC cells in vitro and in vivo by suppressing the expression of KIAA1199 and several key proteins in the Wnt/ß-catenin and EGFR signaling pathways (e.g., WBP11, FGFR4, and PTP4A3). Taken together, these data demonstrate that KIAA1199 promotes GC metastasis by activating EMT-related signaling pathways and that miR-29c-3p regulates GC cell migration in vitro and in vivo by regulating KIAA1199 expression and activating the FGFR4/Wnt/ß-catenin and EGFR signaling pathways. These findings provide a new understanding of GC development and progression and may provide novel therapeutic strategies for GC.


Subject(s)
DNA-Binding Proteins/metabolism , MicroRNAs/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Proteins/genetics , Stomach Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronoglucosaminidase , Lymphatic Metastasis , Male , Mice , Neoplasm Transplantation , RNA Splicing Factors , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Up-Regulation , Wnt Signaling Pathway
11.
Clin Immunol ; 194: 1-8, 2018 09.
Article in English | MEDLINE | ID: mdl-29906512

ABSTRACT

Increasing rates of life-threatening infections and decreasing susceptibility to antibiotics urge an effective vaccine targeting Staphylococcus aureus. Here we investigate the role of cellular immunity in FnBPA110-263 mediated protection in Staphylococcus aureus infection. This study revealed FnBPA110-263 broadly protected mice from seven FnBPA isotypes strains in the sepsis model. FnBPA110-263 immunized B-cell deficient mice were protected against lethal challenge, while T-cell deficient mice were not. Reconstituting mice with FnBPA110-263 specific CD4+ T-cells conferred antigen specific protection. In vitro assays indicated that isolated FnBPA110-263 specific splenocytes from immunized mice produced abundant IL-17A. IL-17A deficient mice were not protected from a lethal challenge by FnBPA110-263 vaccination. Moreover, neutralizing IL-17A, but not IFN-γ,reverses FnBPA110-263-induced protective efficacy in sepsis and skin infection model. These findings suggest that IL-17A producing Th17 cells play an essential role in FnBPA110-263 vaccine-mediated defense against S. aureus sepsis and skin infection in mice.


Subject(s)
Adhesins, Bacterial/immunology , Bacterial Vaccines/immunology , Sepsis/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Immunity, Cellular/immunology , Interferon-gamma/immunology , Interleukin-17/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Sepsis/microbiology , Th17 Cells/immunology , Th17 Cells/microbiology , Vaccination/methods
12.
Cell Death Dis ; 9(5): 533, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29748623

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) is a frequent glomerular disease, and is the common cause of nephrotic syndrome. However, there is no validated diagnostic blood biomarker for FSGS. Here, we performed a real-time PCR-based high-throughput miRNA profiling to identify the plasma signature for FSGS. We found four miRNAs (miR-17, miR-451, miR-106a, and miR-19b) were significantly downregulated in the plasma of FSGS patients (n = 97) compared with healthy controls (n = 124) in the training, validation, and blinded-test phases. The miRNA panel produced an AUC value of 0.82, and was associated with FSGS severity and histologic classification. A three-miRNA panel, including miR-17, miR-451, and miR-106a was related to FSGS remission. Furthermore, the downregulation of plasma-miRNA signature was not detected in disease controls (n = 119) such as IgA nephropathy (IgAN), mesangial proliferative glomerulonephritis (MSPGN), and membranous nephropathy (MN), and the miRNA panel discriminated between FSGS and disease controls. Pathway analysis showed that the four-miRNA panel may cooperatively regulate the pathways involved in the development of FSGS, such as apoptosis. We identified that phosphatase and tensin homolog (PTEN), Bcl-2-like protein 11 (BCL2L11), and chemokine (C-X-C motif) ligand 14 (CXCL14) were targets of miR-106a in human podocyte. Additionally, miR-106a overexpression suppressed podocyte apoptosis in vitro and the downregulation of four-miRNA panel probably resulted in the enhanced apoptosis in podocyte during FSGS development. Taken together, our data show that the plasma-miRNA panel is a potential independent diagnostic and prognostic factor for FSGS. Above miRNAs are involved in FSGS pathogenesis through regulating podocyte apoptosis.


Subject(s)
Apoptosis , Glomerulosclerosis, Focal Segmental/blood , MicroRNAs/blood , Podocytes/metabolism , Biomarkers/blood , Female , Glomerulosclerosis, Focal Segmental/pathology , Humans , Male , Podocytes/pathology
13.
Cancer Sci ; 109(5): 1393-1403, 2018 May.
Article in English | MEDLINE | ID: mdl-29569792

ABSTRACT

Gastric cancer (GC) is among the most fatal cancers in China. MicroRNAs (miRNAs) are versatile regulators during GC development and progression. miR-491-5p has been demonstrated to act as a tumor suppressor in several types of cancer. However, the role of miR-491-5p in GC metastasis remains unknown. Here, we found that miR-491-5p was significantly decreased in GC tissues compared with adjacent non-cancerous tissues, and low miR-491-5p level was associated with large tumor size. Overexpression of miR-491-5p significantly suppressed GC cell epithelial-to-mesenchymal transition (EMT) and tumor metastasis in vitro and in vivo. Mechanistically, SNAIL was identified as a direct target of miR-491-5p. The silencing of SNAIL phenocopied the tumor suppressive function of miR-491-5p, whereas re-expression of SNAIL in GC cells rescued the EMT markers and cell migratory ability that were inhibited by miR-491-5p. In addition, miR-491-5p inhibited FGFR4 indirectly. Inhibition of FGFR4 also decreased the SNAIL level and impaired EMT and cell migration. Taken together, these findings indicate that downregulation of miR-491-5p promoted GC metastasis by inducing EMT via regulation of SNAIL and FGFR4.


Subject(s)
MicroRNAs/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Snail Family Transcription Factors/genetics , Stomach Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Down-Regulation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Snail Family Transcription Factors/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Burden
14.
Clin Immunol ; 188: 85-93, 2018 03.
Article in English | MEDLINE | ID: mdl-29289748

ABSTRACT

With more and more drug-resistant Staphylococcus aureus strains emerging in hospitals, there is an urgent need to develop an effective vaccine to combat S. aureus infection. In this study, we constructed a novel bivalent fusion vaccine, SpA-DKKAA-FnBPA37-507 (SF), based on the D domain of staphylococcal protein A (SpA) and the A domain of fibronectin-binding protein A (FnBPA). Immunisation with SF induced a more ideal protective effect compared with the single components alone in a sepsis model. It also showed broad immunoprotection against seven FnBPA isotypes. Vaccination with SF induced strong antibodies responses and Th1/Th17 polarized cellular responses. Further we demonstrated the protective effect of antibodies by the opsonophagocytic assay (OPA) and passive immunisation. Moreover, vaccination with SF showed protective efficacy in a murine pneumonia model and skin abscess model. These results suggest that SF can be regarded as a promising vaccine candidate for the prevention of S. aureus infections.


Subject(s)
Disease Models, Animal , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Vaccines/immunology , Animals , Antibodies, Bacterial/immunology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Immunity, Cellular/immunology , Immunization/methods , Mice , Pneumonia/immunology , Pneumonia/microbiology , Pneumonia/prevention & control , Sepsis/immunology , Sepsis/microbiology , Sepsis/prevention & control , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcal Protein A/immunology , Staphylococcal Protein A/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Vaccines/administration & dosage
15.
RSC Adv ; 8(19): 10425-10436, 2018 Mar 13.
Article in English | MEDLINE | ID: mdl-35540467

ABSTRACT

Nanoemulsion adjuvants-based vaccines have potent induced immune responses against methicillin-resistant Staphylococcus aureus (MRSA) infection. However, the efficacies and immune responses of different antigen-attaching ways on self-made nanoemulsion adjuvants remain unknown. In this study, we designed three formulations of nanoemulsion adjuvants (encapsulation, mixture, and combination) to explore their immune response-enhancing effects and their underlying mechanism in a systemic infection model of MRSA. Our results showed that the three nanoemulsion-attachment ways formulated with a fusion antigen of MRSA (HlaH35LIsdB348-465) all improved humoral and cellular immune responses. When compared with the mixture and combination formulations, the nanoemulsion-encapsulation group effectively promoted the antigen uptake of dendritic cells (DCs) in vitro, the activation of DC in draining lymph nodes and the delayed release of antigen at injection sites in vivo. Moreover, the encapsulation group induced a more ideal protective efficacy in a MRSA sepsis model by inducing more potent antibody responses and a Th1/Th17 biased CD4+ T cell response when compared with the other two attachment ways. Our findings suggested that the encapsulated formulation of vaccine with nanoemulsion adjuvant is an effective attachment way to provide protective immunity against MRSA infection.

16.
Sci Rep ; 6: 30923, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27484289

ABSTRACT

T lymphocyte-mediated immune responses are critical for antitumour immunity; however, T cell function is impaired in the tumour environment. MicroRNAs are involved in regulation of the immune system. While little is known about the function of intrinsic microRNAs in CD8(+) T cells in the tumour microenvironment. Here, we found that miR-491 was upregulated in CD8(+) T cells from mice with colorectal cancer. Retroviral overexpression of miR-491 in CD8(+) and CD4(+) T cells inhibited cell proliferation and promoted cell apoptosis and decreased the production of interferon-γ in CD8(+) T cells. We found that miR-491 directly targeted cyclin-dependent kinase 4, the transcription factor T cell factor 1 and the anti-apoptotic protein B-cell lymphoma 2-like 1 in CD8(+) T cells. Furthermore, tumour-derived TGF-ß induced miR-491 expression in CD8(+) T cells. Taken together, our results suggest that miR-491 can act as a negative regulator of T lymphocytes, especially CD8(+) T cells, in the tumour environment; thus, this study provides a novel insight on dysfunctional CD8(+) T cells during tumourigenesis and cancer progression. In conclusion, miR-491 may be a new target for antitumour immunotherapy.


Subject(s)
Apoptosis , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/pathology , MicroRNAs/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Female , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
17.
PLoS One ; 11(2): e0149638, 2016.
Article in English | MEDLINE | ID: mdl-26895191

ABSTRACT

Vaccination strategies for Staphylococcus aureus, particularly methicillin-resistant S. aureus (MRSA) infections have attracted much research attention. Recent efforts have been made to select manganese transport protein C, or manganese binding surface lipoprotein C (MntC), which is a metal ion associated with pathogen nutrition uptake, as potential candidates for an S. aureus vaccine. Although protective humoral immune responses to MntC are well-characterised, much less is known about detailed MntC-specific B cell epitope mapping and particularly epitope vaccines, which are less-time consuming and more convenient. In this study, we generated a recombinant protein rMntC which induced strong antibody response when used for immunisation with CFA/IFA adjuvant. On the basis of the results, linear B cell epitopes within MntC were finely mapped using a series of overlapping synthetic peptides. Further studies indicate that MntC113-136, MntC209-232, and MntC263-286 might be the original linear B-cell immune dominant epitope of MntC, furthermore, three-dimensional (3-d) crystal structure results indicate that the three immunodominant epitopes were displayed on the surface of the MntC antigen. On the basis of immunodominant MntC113-136, MntC209-232, and MntC263-286 peptides, the epitope vaccine for S. aureus induces a high antibody level which is biased to TH2 and provides effective immune protection and strong opsonophagocytic killing activity in vitro against MRSA infection. In summary, the study provides strong proof of the optimisation of MRSA B cell epitope vaccine designs and their use, which was based on the MntC antigen in the development of an MRSA vaccine.


Subject(s)
Bacterial Proteins/immunology , Cation Transport Proteins/immunology , Epitopes, B-Lymphocyte/immunology , Immunodominant Epitopes/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Cation Transport Proteins/genetics , Epitope Mapping , Female , HL-60 Cells , Hemocyanins/immunology , Humans , Manganese , Mice , Mice, Inbred BALB C , Phagocytosis , Staphylococcal Infections/immunology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/genetics , Vaccines, Conjugate/immunology , Vaccines, Synthetic/immunology
18.
Sci Rep ; 6: 20929, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26865417

ABSTRACT

Staphylococcus aureus causes serious sepsis and necrotic pneumonia worldwide. Due to the spread of multidrug-resistant strains, developing an effective vaccine is the most promising method for combating S. aureus infection. In this study, based on the immune-dominant areas of the iron surface determinant B (IsdB) and clumping factor A (ClfA), we designed the novel chimeric vaccine IsdB151-277ClfA33-213 (IC). IC formulated with the AlPO4 adjuvant induced higher protection in an S. aureus sepsis model compared with the single components alone and showed broad immune protection against several clinical S. aureus isolates. Immunisation with IC induced strong antibody responses. The protective effect of antibodies was demonstrated through the opsonophagocytic assay (OPA) and passive immunisation experiment. Moreover, this new chimeric vaccine induced Th1/Th17-skewed cellular immune responses based on cytokine profiles and CD4(+) T cell stimulation tests. Neutralisation of IL-17A alone (but not IFN-γ) resulted in a significant decrease in vaccine immune protection. Finally, we found that IC showed protective efficacy in a pneumonia model. Taken together, these data provide evidence that IC is a potentially promising vaccine candidate for combating S. aureus sepsis and pneumonia.


Subject(s)
Antibodies, Bacterial/biosynthesis , Pneumonia/prevention & control , Recombinant Fusion Proteins/administration & dosage , Sepsis/prevention & control , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cation Transport Proteins/genetics , Cation Transport Proteins/immunology , Cloning, Molecular , Coagulase/genetics , Coagulase/immunology , Disease Models, Animal , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Expression , Humans , Immunization , Interleukin-17/antagonists & inhibitors , Interleukin-17/genetics , Interleukin-17/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/microbiology , Pneumonia/mortality , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sepsis/immunology , Sepsis/microbiology , Sepsis/mortality , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/mortality , Staphylococcal Vaccines/biosynthesis , Staphylococcal Vaccines/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity , Survival Analysis
19.
Dig Dis Sci ; 61(1): 80-90, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26289258

ABSTRACT

BACKGROUND: Vacuolating cytotoxin antigen (VacA) is one of the major virulence factors in Helicobacter pylori (H. pylori), which is responsible for cell vacuolar degeneration and apoptotic cell death. A candidate host factor which mediates this process is cortactin, a protein associated with the processes of colonization and adhesion of H. pylori in gastric epithelium. AIM: To investigate the role of cortactin in VacA-induced apoptosis of gastric epithelial cells. METHODS: Cortactin expression and shRNA lentiviral constructs were developed and transduced into the human gastric cancer cell line, AGS. VacA protein was purified from H. pylori cultures, acid-activated, and co-incubated with the transduced cell populations. Apoptosis was detected by flow cytometry, and the levels of the pro- and anti-apoptotic proteins Bax and Bcl-2 were determined by Western blot. RESULTS: Acid-activated purified VacA induced apoptosis in the parental AGS cells. Increased expression of cortactin (AGS/cortactin) led to a greater percentage of cells undergoing apoptosis. In contrast, knockdown of cortactin with shRNA (AGS/cortactin-shRNA) decreased the percentage of apoptotic cells. The protein levels of pro- and anti-apoptotic proteins Bax and Bcl-2 were increased and decreased in AGS/cortactin cells relative to the parental AGS cells. In the AGS/cortactin-shRNA cells, Bax protein levels were decreased, while Bcl-2 protein was increased. CONCLUSIONS: The results indicate that cortactin is involved in the regulation of apoptosis induced by VacA in gastric cells.


Subject(s)
Apoptosis , Bacterial Proteins/metabolism , Cortactin/metabolism , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Cell Line, Tumor , Cortactin/genetics , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Signal Transduction , Transduction, Genetic , Transfection , bcl-2-Associated X Protein/metabolism
20.
Int J Nanomedicine ; 10: 7275-90, 2015.
Article in English | MEDLINE | ID: mdl-26664118

ABSTRACT

The Gram-positive bacterial pathogen methicillin-resistant Staphylococcus aureus (MRSA) can cause infections in the bloodstream, endocardial tissue, respiratory tract, culture-confirmed skin, or soft tissue. There are currently no effective vaccines, and none are expected to become available in the near future. An effective vaccine capable of eliciting both systemic and mucosal immune responses is also urgently needed. Here, we reported a novel oil-in-water nanoemulsion adjuvant vaccine containing an MRSA recombination protein antigen, Cremophor EL-35(®) as a surfactant, and propylene glycol as a co-surfactant. This nanoemulsion vaccine, whose average diameter was 31.34±0.49 nm, demonstrated good protein structure integrity, protein specificity, and good stability at room temperature for 1 year. The intramuscular systemic and nasal mucosal immune responses demonstrated that this nanoemulsion vaccine could improve the specific immune responses of immunoglobulin (Ig)G and related subclasses, such as IgG1, IgG2a, and IgG2b, as well as IgA, in the serum after Balb/c mice intramuscular immunization and C57 mice nasal immunization. Furthermore, this nanoemulsion vaccine also markedly enhanced the interferon-γ and interleukin-17A cytokine cell immune response, improved the survival ratio, and reduced bacterial colonization. Taken together, our results show that this novel nanoemulsion vaccine has great potential and is a robust generator of an effective intramuscular systemic and nasal mucosal immune response without the need for an additional adjuvant. Thus, the present study serves as a sound scientific foundation for future strategies in the development of this novel nanoemulsion adjuvant vaccine to enhance both the intramuscular systemic and nasal mucosal immune responses.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity, Mucosal/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Nanoparticles/chemistry , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Animals , Emulsions , Female , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice, Inbred BALB C , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , Static Electricity , Survival Analysis , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...