Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
1.
Bull Exp Biol Med ; 169(4): 467-469, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32910376

ABSTRACT

Cultured peritoneal macrophages from intact (control) and BCG-infected (experiment) male BALB/c mice were studied 90 days after infection. Polarization of macrophages by M1 (expression of GM-CSF, IFNγ, and CD16/32) and M2 (expression of bFGF and CD36) differentiation pathways was studied with consideration for their the nuclearity class. Mononuclear cells predominated (90% and higher) in macrophage cultures of both groups and presumably, were presented by mainly epithelioid cells. The results indicated polarization of mononuclear and multinuclear macrophages in the M2 direction under conditions of BCG granulomatosis and a higher initial M2 polarization of binuclear macrophages. In control cultures, the ratio of M2 to M1 macrophages was 0.57, in experimental cultures this ratio was 1.6. It seems that long persistence of Mycobacterium tuberculosis in macrophages served as a factor stimulating the plastic processes and transformation of macrophages into epithelioid cells that form the "core" of granulomas and their enlargement upon incorporation of macrophages.


Subject(s)
Epithelioid Cells/pathology , Gene Expression Regulation/immunology , Macrophages, Peritoneal/pathology , Mycobacterium bovis/growth & development , Tuberculosis/pathology , Animals , CD36 Antigens/genetics , CD36 Antigens/immunology , Cell Differentiation , Cell Transdifferentiation/genetics , Cell Transdifferentiation/immunology , Epithelioid Cells/immunology , Epithelioid Cells/microbiology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , Male , Mice , Mice, Inbred BALB C , Mycobacterium bovis/pathogenicity , Primary Cell Culture , Receptors, IgG/genetics , Receptors, IgG/immunology , Tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis/microbiology
2.
Int J Mol Sci ; 21(15)2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32731615

ABSTRACT

Mesenchymal stem cells (MSCs) have become a promising tool in cellular therapy for restoring immune system haemostasis; however, the success of clinical trials has been impaired by the lack of standardized manufacturing processes. This study aims to determine the suitability of source tissues and culture media for the production of MSC-based advanced therapy medicinal products (ATMPs) and to define parameters to extend the set of release criteria. MSCs were isolated from umbilical cord (UC), bone marrow and lipoaspirate and expanded in three different culture media. MSC phenotype, proliferation capacity and immunosuppressive parameters were evaluated in normal MSCs compared to primed MSCs treated with cytokines mimicking an inflammatory environment. Compared to bone marrow and lipoaspirate, UC-derived MSCs (UC-MSCs) showed the highest proliferative capacity, which was further enhanced by media supplemented with bFGF, while the cells maintained their immunosuppressive characteristics. Moreover, UC-MSCs expanded in the bFGF-enriched medium were the least sensitive to undesirable priming-induced changes in the MSC phenotype. Surface markers and secreted factors were identified to reflect the cell response to inflammatory priming and to be variable among MSCs from different source tissues. This study demonstrates that UC is a favorable cell source for manufacturing MSC-based ATMPs for immunosuppressive applications. UC-MSCs are able to use the bFGF-enriched medium for higher cell yields without the impairment of immunosuppressive parameters and undesirable phenotype changes after inflammatory preconditioning of MSCs before transplantation. Additionally, immunosuppressive parameters were identified to help finding predictors of clinically efficient MSCs in the following clinical trials.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Fibroblast Growth Factor 2/pharmacology , Immunosuppression Therapy , Mesenchymal Stem Cells/immunology , Umbilical Cord/immunology , Cell Differentiation/immunology , Fibroblast Growth Factor 2/immunology , Humans , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology
3.
Nanomedicine ; 29: 102254, 2020 10.
Article in English | MEDLINE | ID: mdl-32615335

ABSTRACT

FGF-2 accumulates in many tumor tissues and is closely related to the development of tumor angiogenesis and the immunosuppressive microenvironment. This study aimed to investigate whether active immunization against FGF-2 could modify antitumor immunity and enhance the efficacy of an HPV16 E7-specific therapeutic vaccine. Combined immunization targeting both FGF-2 and E7 significantly suppressed tumor growth, which was accompanied by significantly increased levels of IFN-γ-expressing splenocytes and effector CD8 T cells and decreased levels of immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells(MDSCs) in both the spleen and tumor; in addition, the levels of FGF-2 and neovascularization in tumors were decreased in the mice receiving the combined immunization, and tumor cell apoptosis was promoted. The combination of an HPV16 E7-specific vaccine and active immunization against FGF-2 significantly enhances antitumor immune responses in mice with TC-1 tumors, indicating a promising strategy for tumor immunotherapy.


Subject(s)
Cancer Vaccines/pharmacology , Fibroblast Growth Factor 2/immunology , Neovascularization, Pathologic/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/pharmacology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/genetics , Humans , Immunotherapy , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/prevention & control , Neovascularization, Pathologic/virology , Papillomavirus E7 Proteins/antagonists & inhibitors , Papillomavirus E7 Proteins/genetics , Papillomavirus Vaccines/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Vaccination
4.
Int J Nanomedicine ; 15: 1983-1996, 2020.
Article in English | MEDLINE | ID: mdl-32308382

ABSTRACT

BACKGROUND: Fibroblast growth factor (FGF)-2 is overexpressed in various tumor tissues. It affects tumor cell proliferation, invasion and survival, promotes tumor angiogenesis and is tightly involved in the development of systemic and local immunosuppressive tumor mechanisms. PURPOSE: This study aimed to develop an effective vaccine against FGF-2 and to investigate the effects of anti-FGF-2 immunization on tumor growth and antitumor immune responses. METHODS: A set of thirteen synthesized overlapping peptides covering all possible linear B-cell epitopes of murine FGF-2 and a recombinant FGF-2 protein were conjugated to virus-like particles (VLPs) of recombinant hepatitis B core antigen (HBcAg). The VLPs were immunized through a preventive or therapeutic strategy in a TC-1 or 4T1 grafted tumor model. RESULTS: Immunization with FGF-2 peptides or full-length protein-coupled VLPs produced FGF-2-specific antibodies with a high titer. Peptide 12, which is located in the heparin-binding site of FGF-2, or protein-conjugated VLPs presented the most significant effects on the suppression of TC-1 tumor growth. The levels of IFN-γ-expressing splenocytes and serum IFN-γ were significantly elevated; further, the immune effector cells CD8+ IFN-γ+ cytotoxic T lymphocytes (CTLs) and CD4+ IFN-γ+ Th1 cells were significantly increased, whereas the immunosuppressive cells CD4+ CD25+ FOXP3+ Treg cells and Gr-1+ CD11b+ myeloid-derived suppressor cells (MDSCs) were decreased in the immunized mice. In addition, VLP immunization significantly suppressed tumor vascularization and promoted tumor cell apoptosis. In mice bearing 4T1 breast tumor, preventive immunization with FGF-2-conjugated VLPs suppressed tumor growth and lung metastasis, and increased effector cell responses. CONCLUSION: Active immunization against FGF-2 is a new possible strategy for tumor immunotherapy.


Subject(s)
Cancer Vaccines/pharmacology , Epitopes, B-Lymphocyte/immunology , Fibroblast Growth Factor 2/immunology , Peptides/immunology , Vaccines, Virus-Like Particle/pharmacology , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Female , Hepatitis B Core Antigens/genetics , Hepatitis B Core Antigens/immunology , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunotherapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Neovascularization, Pathologic/drug therapy , Peptides/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Vaccination , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology
5.
Exp Eye Res ; 190: 107823, 2020 01.
Article in English | MEDLINE | ID: mdl-31600485

ABSTRACT

Administration of RC28-E, a VEGF/bFGF dual decoy receptor (IgG1 Fc-fusion protein), have shown relative therapeutic value in ocular in vivo models, including laser-induced choroidal neovascularization (CNV) in monkeys and streptozotocin (STZ)-induced diabetic retinopathy (DR) in rats. In the present study, we have elucidated the pharmacokinetics profiles of RC28-E at the systemic, vitreous and aqueous humor after administration in a primate model (Macaca fascicularis). Moreover, here we tease out the ocular tissue distribution of RC28-E after intravitreal administration, and we also determine the systemic bioavailability after both intravitreal and intravenous administration. Our results show that RC28-E is rapidly and well-distributed into ocular tissues after intravitreal administration. Drug exposure in choroid and retina was approximately one-quarter and one-twelfth of that in vitreous humor, while its half-life in vitreous and aqueous humor were well-sustained (3.3 and 3.0 days). Remarkably, RC28-E could cross the blood-ocular barrier, and the systemic bioavailability of RC28-E was ~25%. No drug accumulation after multiple administration was noticed, but low titers of antibody produce against RC28-E were detected. Overall, RC28-E exhibited high clinical value due to adequate pharmacokinetic profiling, safety and efficacy.


Subject(s)
Aqueous Humor/metabolism , Fibroblast Growth Factor 2/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Vascular Endothelial Growth Factor A/pharmacokinetics , Vitreous Body/metabolism , Animals , Biological Availability , Blood-Retinal Barrier , Choroid/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fibroblast Growth Factor 2/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Intravitreal Injections , Lens, Crystalline/metabolism , Macaca fascicularis , Male , Recombinant Fusion Proteins/immunology , Retina/metabolism , Tissue Distribution , Vascular Endothelial Growth Factor A/immunology
6.
J Biochem ; 165(6): 487-495, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30597085

ABSTRACT

The human fibroblast growth factor-2 (FGF-2) highly expressed in tumours is an important factor to promote tumour angiogenesis and lymphangiogenesis. A disulphide-stabilized diabody (ds-Diabody) could specifically target FGF-2 and show its advantages in inhibition of tumour angiogenesis and growth. It is very important for antibody drugs to confirm the fine epitope. Here, theoretical structure models of FGF-2 and antibody were built by homology modelling. The amino acid residues in the interaction interface of antigen and antibody were analysed by molecular docking. The potential epitope was predicted by homology modelling and molecular docking of antigen-antibody and site-directed mutation assays of alanine scanning. The predicted epitope was verified by antigen mutagenesis and enzyme-linked immunosorbent assay (ELISA). The epitope mapping assay showed that the epitope of ds-Diabody against FGF-2 was defined by the discontinuous sites including six amino acid residues (P23, Q65, R69, G70, Y82 and R118). The results showed that the epitope was localized in the interaction interface of FGF-2 and ds-Diabody. The fine epitope mapping provided the important information for understanding the inhibition activity of ds-Diabody against FGF-2 and helping in the further development of ds-Diabody against FGF-2 as a potentially promising antibody drug for future cancer therapy.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Disulfides/chemistry , Epitope Mapping , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/immunology , Neoplasms/drug therapy , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/therapeutic use , Antigen-Antibody Reactions , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2/genetics , HEK293 Cells , Humans , Models, Molecular , Mutagenesis, Site-Directed , Neoplasms/immunology , Neoplasms/pathology
7.
Eur Rev Med Pharmacol Sci ; 23(1): 16-22, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30657541

ABSTRACT

OBJECTIVE: The study aimed to investigate the correlations of insulin resistance and hemoglobin A1c (HbA1c) with cytokines [insulin-like growth factor 1 (IGF-1), basic fibroblast growth factor (bFGF) and interleukin-6 (IL-6)] in the aqueous humor of patients with diabetic cataract. PATIENTS AND METHODS: 59 patients with diabetic cataract and 58 patients with simple cataract treated in Jining No. 1 People´s Hospital (Jining, China) from January 2017 to February 2018, were selected randomly. The levels of homeostasis model assessment of insulin resistance (HOMA-IR) and HbAlc, as well as IGF-1, bFGF and IL-6 in the aqueous humor were compared between the two groups. The correlations of HOMA-IR and HbAlc with IGF-1, bFGF and IL-6 were analyzed. In control group, the levels of HOMA-IR and HbAlc, as well as IGF-1, bFGF and IL-6 in the aqueous humor were significantly lower than those in observation group (p<0.05). RESULTS: Compared with the group with HbAlc ≤ 7%, the groups with HbAlc ≥ 9% and 7%

Subject(s)
Aqueous Humor/chemistry , Cataract/diagnosis , Diabetes Complications/diagnosis , Glycated Hemoglobin/analysis , Insulin Resistance , Adult , Biomarkers/analysis , Case-Control Studies , Cataract/blood , Cataract/immunology , Diabetes Complications/blood , Diabetes Complications/immunology , Female , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/immunology , Humans , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/immunology , Interleukin-6/analysis , Interleukin-6/immunology , Male , Middle Aged , Predictive Value of Tests
8.
J Biol Chem ; 293(44): 17218-17228, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30217817

ABSTRACT

Fibroblast growth factor receptor-1 (FGFR1) activity at the plasma membrane is tightly controlled by the availability of co-receptors and competing receptor isoforms. We have previously shown that FGFR1 activity in pancreatic beta-cells modulates a wide range of processes, including lipid metabolism, insulin processing, and cell survival. More recently, we have revealed that co-expression of FGFR5, a receptor isoform that lacks a tyrosine-kinase domain, influences FGFR1 responses. We therefore hypothesized that FGFR5 is a co-receptor to FGFR1 that modulates responses to ligands by forming a receptor heterocomplex with FGFR1. We first show here increased FGFR5 expression in the pancreatic islets of nonobese diabetic (NOD) mice and also in mouse and human islets treated with proinflammatory cytokines. Using siRNA knockdown, we further report that FGFR5 and FGFR1 expression improves beta-cell survival. Co-immunoprecipitation and quantitative live-cell imaging to measure the molecular interaction between FGFR5 and FGFR1 revealed that FGFR5 forms a mixture of ligand-independent homodimers (∼25%) and homotrimers (∼75%) at the plasma membrane. Interestingly, co-expressed FGFR5 and FGFR1 formed heterocomplexes with a 2:1 ratio and subsequently responded to FGF2 by forming FGFR5/FGFR1 signaling complexes with a 4:2 ratio. Taken together, our findings identify FGFR5 as a co-receptor that is up-regulated by inflammation and promotes FGFR1-induced survival, insights that reveal a potential target for intervention during beta-cell pathogenesis.


Subject(s)
Cytokines/immunology , Diabetes Mellitus/genetics , Insulin-Secreting Cells/immunology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Animals , Diabetes Mellitus/immunology , Dimerization , Female , Fibroblast Growth Factor 2/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/immunology , Receptor, Fibroblast Growth Factor, Type 5/chemistry , Receptor, Fibroblast Growth Factor, Type 5/immunology , Up-Regulation
9.
Eur Rev Med Pharmacol Sci ; 22(6): 1554-1568, 2018 03.
Article in English | MEDLINE | ID: mdl-29630097

ABSTRACT

OBJECTIVE: We aimed at exploring the positive feedback loop in eutopic and ectopic endometrial glandular epithelial cells (EuECs and EECs) in endometriosis. MATERIALS AND METHODS: Normal epithelial cells (NECs), EuECs and EECs were treated with fibroblast growth factor (FGF)2, FGF2 neutralizing antibody, mitogen-activated protein kinases (MAPKs) inhibitors U0126 and PD98059. FGF2 protein level was detected by enzyme-linked immunosorbent assay (ELISA). The expressions of FGF2, FGF receptor 1 (FGFR1), extracellular signal-regulated kinase (ERK)1/2/pERK1/2 and Sproutys (SPRYs) (Sprouty1, Sprouty2, Sprouty4) and dual specificity phosphatase 6 (DUSP6) were detected by Western blot. The mRNA levels of FGF2, FGFR1 (FGF receptor 1), SPRYs (Sprouty1, Sprouty2, Sprouty4) and DUSP6 mRNA were detected by RT-PCR. RESULTS: Among treatment groups, the content of FGF2 in EuECs and EECs was significantly higher than that in NECs (p < 0.05). The mRNA and protein levels of FGF2, FGFR1, SPRYs (Sprouty1, Sprouty2, Sprouty4) and DUSP6 in EuECs and EECs were increased after adding FGF2 (p < 0.05), but decreased after adding FGF2 neutralizing antibody, no significant change was found in NECs (p > 0.05). The inhibitory effect of PD9805 on NECs was not significantly different from that of U0126 (p > 0.05); however, the inhibitory effects of PD9805 on EuECs and EECs were significantly lower than those of U0126 (p< 0.05). CONCLUSIONS: The positive feedback loop existed in EuECs and EECs, but maybe not in NECs. The results may provide the guideline to treat endometriosis patients.


Subject(s)
Endometriosis/pathology , Fibroblast Growth Factor 2/pharmacology , Signal Transduction/drug effects , Antibodies, Neutralizing/pharmacology , Butadienes/pharmacology , Cell Line , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/metabolism , Endometriosis/metabolism , Endometrium/cytology , Endometrium/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Fibroblast Growth Factor 2/immunology , Fibroblast Growth Factor 2/metabolism , Flavonoids/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nitriles/pharmacology , Phosphoproteins/genetics , Phosphoproteins/metabolism
10.
Bull Exp Biol Med ; 164(4): 456-458, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29504104

ABSTRACT

The incidence of mono- and multinuclear cells and their expression of pro- and antifibrotic factors were studied in cultured peritoneal macrophages from intact and BCG-infected mice. Generally, the expression of factors increased with an increase in the number of nuclei per cell. However, the expression was higher in macrophages from BCG infected mice, except the cells with 3 and more nuclei, extremely rarely expressing IL-1α in cultures from intact and BCG-infected animals. The number of macrophages with 3 and more nuclei, expressing CatD, was comparable with the number of mono- and binuclear macrophages. Presumably, this was determined by various mechanisms of formation of multinuclear (3-5 and more nuclei) macrophages, for example, by amitosis.


Subject(s)
Cathepsin D/immunology , Fibroblast Growth Factor 2/immunology , Interferon-gamma/immunology , Interleukin-1alpha/immunology , Macrophages, Peritoneal/immunology , Matrix Metalloproteinase 13/immunology , Mycobacterium Infections/immunology , Transforming Growth Factor beta1/immunology , Animals , Cathepsin D/genetics , Cell Nucleus , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation , Interferon-gamma/genetics , Interleukin-1alpha/genetics , Macrophages, Peritoneal/microbiology , Macrophages, Peritoneal/pathology , Male , Matrix Metalloproteinase 13/genetics , Mice , Mice, Inbred BALB C , Mycobacterium Infections/microbiology , Mycobacterium Infections/pathology , Mycobacterium bovis/immunology , Primary Cell Culture , Signal Transduction , Transforming Growth Factor beta1/genetics
11.
Cancer Biol Ther ; 19(6): 518-524, 2018 06 03.
Article in English | MEDLINE | ID: mdl-29405828

ABSTRACT

Tumor growth and metastasis are closely related to angiogenesis. Basic fibroblast growth factor(bFGF) is an angiogenic factor, and up-regulated expression of bFGF plays a crucial role in the development and metastasis of melanoma. Therefore, in this study, we sought to achieve antitumor activity by immunity targeting bFGF which would inhibit tumor angiogenesis and simultaneously induce bFGF specific cytotoxic T lymphocytes to kill melanoma cells. A human bFGF protein was used as exogenous antigen, coupled with a saponin-liposome adjuvant formulation to enhance CTL response. The results showed that the immunity induced strong immune response and produced prominent anti-cancer activities. CD31 immunohistochemistry and alginate-encapsulated tumor cell assay displayed that tumor angiogenesis was effectively inhibited. Further, the higher production of IFN-γ and cytotoxic T lymphocyte killing assay suggested that the anti-cancer activities may mainly depend on cellular immune response, which could cause the inhibition of tumor angiogenesis and specific killing of tumor cells by bFGF-specific cytotoxic T lymphocytes. We concluded that immunotherapy targeting bFGF may be a prominent strategy for melanoma, and that the adjuvant formulation of saponin-liposome is very desirable in enhancing cytotoxic T lymphocytes response.


Subject(s)
Cancer Vaccines/therapeutic use , Fibroblast Growth Factor 2/immunology , Immunotherapy/methods , Melanoma/drug therapy , Animals , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Disease Models, Animal , Female , Humans , Melanoma/pathology , Mice
12.
J Mol Cell Biol ; 10(6): 573-585, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29121325

ABSTRACT

Influenza virus (IAV) infection is a major cause of severe respiratory illness that affects almost every country in the world. IAV infections result in respiratory illness and even acute lung injury and death, but the underlying mechanisms responsible for IAV pathogenesis have not yet been fully elucidated. In this study, the basic fibroblast growth factor 2 (FGF2) level was markedly increased in H1N1 virus-infected humans and mice. FGF2, which is predominately derived from epithelial cells, recruits and activates neutrophils via the FGFR2-PI3K-AKT-NFκB signaling pathway. FGF2 depletion or knockout exacerbated influenza-associated disease by impairing neutrophil recruitment and activation. More importantly, administration of the recombinant FGF2 protein significantly alleviated the severity of IAV-induced lung injury and promoted the survival of IAV-infected mice. Based on the results from experiments in which neutrophils were depleted and adoptively transferred, FGF2 protected mice against IAV infection by recruiting neutrophils. Thus, FGF2 plays a critical role in preventing IAV-induced lung injury, and FGF2 is a promising potential therapeutic target during IAV infection.


Subject(s)
Acute Lung Injury/virology , Fibroblast Growth Factor 2/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Neutrophil Infiltration , Orthomyxoviridae Infections/immunology , Acute Lung Injury/etiology , Acute Lung Injury/immunology , Animals , Female , Fibroblast Growth Factor 2/therapeutic use , Humans , Influenza, Human/complications , Influenza, Human/therapy , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/therapy , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use
13.
Nan Fang Yi Ke Da Xue Xue Bao ; 37(11): 1535-1539, 2017 Nov 20.
Article in Chinese | MEDLINE | ID: mdl-29180337

ABSTRACT

OBJECTIVE: To study the synergistic inhibitory effects of basic fibroblast growth factor (bFGF) monoclonal antibody (bFGF mAb) and irinotecan on the proliferation of small cell lung cancer H223 cells. METHODS: CCK-8 assay and flow cytometry were used to assess the effects of bFGF mAb combined with irinotecan on the proliferation and apoptosis of H223 cells, respectively. Western blotting was performed to analyze the effect of bFGF-mAb combined with irinotecan on AKT and ERK1/2 phosphorylation in the cells. RESULTS: Both bFGF mAb and irinotecan alone inhibited H223 cell proliferation in a dose-dependent manner (P<0.05). The inhibitory rate was significantly higher in H223 cells treated with bFGF mAb + irinotecan (54.30%) than in cell treated with bFGF mAb (18.73%) or irinotecan (21.96%) alone (P<0.05). Both bFGF mAb and irinotecan induced H223 cell apoptosis in a dose-dependent manner (P<0.05), and the combined treatment resulted in a significantly higher early apoptosis rates (6.5%) than treatment with bFGF mAb (2.7%) or irinotecan (4.3%) alone (P<0.05). bFGF mAb and irinotecan, either alone or in combination, significantly inhibited the levels of p-AKT protein and p-ERK1/2 protein without obviously affecting AKT and ERK1/2 protein levels. CONCLUSION: bFGF mAb and irinotecan produce synergistic inhibitory effects on small cell lung cancer H223 cells by suppressing proliferation and promoting apoptosis of the cells, and can effectively block the MAPK/ERK and PI3K/AKT signaling pathways associated with bFGF.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Fibroblast Growth Factor 2/immunology , Irinotecan/pharmacology , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Signal Transduction , Small Cell Lung Carcinoma/pathology
14.
J Immunol ; 199(9): 3086-3093, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28924004

ABSTRACT

Activated CD4 T cells connect to airway smooth muscle cells (ASMCs) in vitro via lymphocyte-derived membrane conduits (LMCs) structurally similar to membrane nanotubes with unknown intercellular signals triggering their formation. We examined the structure and function of CD4 T cell-derived LMCs, and we established a role for ASMC-derived basic fibroblast growth factor 2 (FGF2b) and FGF receptor (FGFR)1 in LMC formation. Blocking FGF2b's synthesis and FGFR1 function reduced LMC formation. Mitochondrial flux from ASMCs to T cells was partially FGF2b and FGFR1 dependent. LMC formation by CD4 T cells and mitochondrial transfer from ASMCs was increased in the presence of asthmatic ASMCs that expressed more mRNA for FGF2b compared with normal ASMCs. These observations identify ASMC-derived FGF2b as a factor needed for LMC formation by CD4 T cells, affecting intercellular communication.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Cell Surface Extensions/immunology , Fibroblast Growth Factor 2/immunology , Myocytes, Smooth Muscle/immunology , CD4-Positive T-Lymphocytes/cytology , Humans , Mitochondria/immunology , Myocytes, Smooth Muscle/cytology , Receptor, Fibroblast Growth Factor, Type 1/immunology , Respiratory System/cytology , Respiratory System/immunology
15.
Oncotarget ; 8(12): 20187-20197, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28423625

ABSTRACT

The over-expression of basic fibroblast growth factor (bFGF) plays a crucial role in the development, invasion and metastasis of lung cancer. Therefore, neutralizing antibodies against bFGF may inhibit the growth of lung cancer. In this study, a Disulfide-stabilized diabody (ds-Diabody) against bFGF was constructed by site-directed mutation and overlap extension PCR (SOE-PCR) at the position of VH44 and VL100 in the scFv. The ds-Diabody was constructed and expressed in Pichia pastoris. We found that the ds-Diabody against bFGF could efficiently suppress the proliferation, migration and invasion of human lung cancer A549 cells in vitro. Moreover, in A549 cells, the ds-Diabody against bFGF could inhibit bFGF-induced activation of downstream signaling regulators, such as phospho-Akt and phospho-MAPK. In the nude mouse xenograft model of lung cancer, the ds-Diabody against bFGF could significantly inhibit tumor growth and decrease the densities of micro-vessels and lymphatic vessels in tumor tissue. Our data indicate that the ds-Diabody against bFGF could effectively suppress the lung cancer growth through blockade of bFGF signaling pathway and inhibition of tumor angiogenesis, which may make it a potential therapeutic candidate antibody drug for human lung cancer therapy.


Subject(s)
Disulfides/chemistry , Fibroblast Growth Factor 2/antagonists & inhibitors , Immunoglobulin Variable Region/pharmacology , Lung Neoplasms/therapy , Neovascularization, Pathologic/prevention & control , Single-Chain Antibodies/pharmacology , Animals , Apoptosis/drug effects , Biomarkers, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Fibroblast Growth Factor 2/immunology , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Respir Res ; 18(1): 48, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28298222

ABSTRACT

Mast cells are accumulated in advanced chronic obstructive pulmonary disease (COPD), and interleukin (IL)-17 signaling plays a role in disease progression. The expression, localization and functional relevance of IL-17 receptor (R)A and IL-17RC was explored in COPD by immunodetection, and functional assays.IL-17RA and IL-17RC was increased in very severe COPD, and expressed by mast cells. Increased secretion of the pro-angiogenic basic fibroblast growth factor and vascular endothelial growth factor was observed in vitro-maintained mast cells stimulated with IL-17A. Expression of these mediators was confirmed in end-stage COPD. Thus, accumulation of mast cells in COPD may contribute to vascular remodeling.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Lung/immunology , Mast Cells/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , Receptors, Interleukin-17/immunology , Receptors, Interleukin/immunology , Vascular Endothelial Growth Factor A/immunology , Aged , Female , Fibroblast Growth Factor 2/immunology , Humans , Male , Mast Cells/immunology , Middle Aged , Up-Regulation/immunology
17.
Transl Psychiatry ; 7(3): e1065, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28323284

ABSTRACT

A subset of patients with depression have elevated levels of inflammatory cytokines, and some studies demonstrate interaction between inflammatory factors and treatment outcome. However, most studies focus on only a narrow subset of factors in a patient sample. In the current study, we analyzed broad immune profiles in blood from patients with treatment-resistant depression (TRD) at baseline and following treatment with the glutamate modulator ketamine. Serum was analyzed from 26 healthy control and 33 actively depressed TRD patients free of antidepressant medication, and matched for age, sex and body mass index. All subjects provided baseline blood samples, and TRD subjects had additional blood draw at 4 and 24 h following intravenous infusion of ketamine (0.5 mg kg-1). Samples underwent multiplex analysis of 41 cytokines, chemokines and growth factors using quantitative immunoassay technology. Our a priori hypothesis was that TRD patients would show elevations in canonical pro-inflammatory cytokines; analyses demonstrated significant elevation of the pro-inflammatory cytokine interleukin-6. Further exploratory analyses revealed significant regulation of four additional soluble factors in patients with TRD. Several cytokines showed transient changes in level after ketamine, but none correlated with treatment response. Low pretreatment levels of fibroblast growth factor 2 were associated with ketamine treatment response. In sum, we found that patients with TRD demonstrate a unique pattern of increased inflammatory mediators, chemokines and colony-stimulating factors, providing support for the immune hypothesis of TRD. These patterns suggest novel treatment targets for the subset of patients with TRD who evidence dysregulated immune functioning.


Subject(s)
Cytokines/immunology , Depressive Disorder, Major/immunology , Depressive Disorder, Treatment-Resistant/immunology , Intercellular Signaling Peptides and Proteins/immunology , Adult , Case-Control Studies , Chemokines/immunology , Depressive Disorder, Major/drug therapy , Depressive Disorder, Treatment-Resistant/drug therapy , Excitatory Amino Acid Antagonists/therapeutic use , Female , Fibroblast Growth Factor 2/immunology , Humans , Inflammation , Infusions, Intravenous , Interleukin-1alpha/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Ketamine/therapeutic use , Male , Middle Aged , Prognosis , Treatment Outcome , Tumor Necrosis Factor-alpha/immunology
18.
Cancer Sci ; 107(8): 1141-50, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27251178

ABSTRACT

Fibroblast growth factor-2 (FGF-2) is one of the most important angiogenic factors to promote tumor growth, progression and metastasis. Neutralizing antibodies against FGF-2 may suppress the growth of tumor cells by blocking the FGF-2 signaling pathway. In this study, a disulfide-stabilized diabody (ds-Diabody) that specifically targets FGF-2 was designed. Compared to its parent antibody, the introduction of disulphide bonds in the diabody could significantly increase the stability of ds-Diabody and maintain its antigen binding activity. The ds-Diabody against FGF-2 could effectively inhibit the tube formation and migration of vascular endothelial cells and block the proliferation and invasion of human breast cancer cells. In the mouse model of breast cancer xenograft tumors, the ds-Diabody against FGF-2 could significantly inhibit the growth of tumor cells. Moreover, the densities of microvessels stained with CD31 and lymphatic vessels stained with LYVE1 in tumors showed a significant decrease following treatment with the ds-Diabody against FGF-2. Our data indicated that the ds-Diabody against FGF-2 could inhibit tumor angiogenesis, lymphangiogenesis and tumor growth.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Disulfides/chemistry , Fibroblast Growth Factor 2/immunology , Immunoglobulins/immunology , Immunoglobulins/pharmacology , Protein Multimerization , Animals , BALB 3T3 Cells , Breast Neoplasms/blood supply , Capillaries/drug effects , Capillaries/growth & development , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Fibroblast Growth Factor 2/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Immunoglobulins/chemistry , Immunoglobulins/isolation & purification , Lymphangiogenesis/drug effects , MCF-7 Cells , Mice , Neoplasm Invasiveness/prevention & control , Neovascularization, Pathologic/drug therapy , Pichia/genetics , Pichia/metabolism , Signal Transduction/drug effects
19.
World J Gastroenterol ; 22(21): 5033-41, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27275095

ABSTRACT

AIM: To investigate the inhibitory efficacy of (125)I-labeled anti-basic fibroblast growth factor (bFGF) monoclonal antibody (mAb) in hepatocellular carcinoma (HCC). METHODS: bFGF mAb was prepared by using the 1G9B9 hybridoma cell line with hybridization technology and extracted from ascites fluid through a Protein G Sepharose affinity column. After labeling with (125)I through the chloramine-T method, bFGF mAb was further purified by a Sephadex G-25 column. Gamma radiation counter GC-1200 detected radioactivity of (125)I-bFGF mAb. The murine H22 HCC xenograft model was established and randomized to interventions with control (phosphate-buffered saline), (125)I-bFGF mAb, (125)I plus bFGF mAb, bFGF mAb, or (125)I. The ratios of tumor inhibition were then calculated. Expression of bFGF, fibroblast growth factor receptor (FGFR), platelet-derived growth factor, and vascular endothelial growth factor (VEGF) mRNA was determined by quantitative reverse transcriptase real-time polymerase chain reaction. RESULTS: The purified bFGF mAb solution was 8.145 mg/mL with a titer of 1:2560000 and was stored at -20 °C. After coupling, (125)I-bFGF mAb was used at a 1: 1280000 dilution, stored at 4 °C, and its specific radioactivity was 37 MBq/mg. The corresponding tumor weight in the control, (125)I, bFGF mAb, (125)I plus bFGF mAb, and (125)I-bFGF mAb groups was 1.88 ± 0.25, 1.625 ± 0.21, 1.5 ± 0.18, 1.41 ± 0.16, and 0.98 ± 0.11 g, respectively. The tumor inhibition ratio in the (125)I, bFGF mAb, (125)I plus bFGF mAb, and (125)I-bFGF mAb groups was 13.6%, 20.2%, 25.1%, and 47.9%, respectively. Growth of HCC xenografts was inhibited significantly more in the (125)I-bFGF mAb group than in the other groups (P < 0.05). Expression of bFGF and FGFR mRNA in the (125)I-bFGF mAb group was significantly decreased in comparison with other groups (P < 0.05). Groups under interventions revealed increased expression of VEGF mRNA (except for (125)I group) compared with the control group. CONCLUSION: (125)I-bFGF mAb inhibits growth of HCC xenografts. The coupling effect of (125)I-bFGF mAb is more effective than the concomitant use of (125)I and bFGF mAb.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carcinoma, Hepatocellular/radiotherapy , Cell Proliferation/radiation effects , Fibroblast Growth Factor 2/immunology , Liver Neoplasms, Experimental/radiotherapy , Radioimmunotherapy/methods , Radiopharmaceuticals/pharmacology , Animals , Antibodies, Monoclonal/metabolism , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Neoplastic , Hybridomas , Iodine Radioisotopes/pharmacology , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...