Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Nat Commun ; 11(1): 4678, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32938916

ABSTRACT

Diabetic foot ulcers (DFUs) are a life-threatening disease that often result in lower limb amputations and a shortened lifespan. However, molecular mechanisms contributing to the pathogenesis of DFUs remain poorly understood. We use next-generation sequencing to generate a human dataset of pathogenic DFUs to compare to transcriptional profiles of human skin and oral acute wounds, oral as a model of "ideal" adult tissue repair due to accelerated closure without scarring. Here we identify major transcriptional networks deregulated in DFUs that result in decreased neutrophils and macrophages recruitment and overall poorly controlled inflammatory response. Transcription factors FOXM1 and STAT3, which function to activate and promote survival of immune cells, are inhibited in DFUs. Moreover, inhibition of FOXM1 in diabetic mouse models (STZ-induced and db/db) results in delayed wound healing and decreased neutrophil and macrophage recruitment in diabetic wounds in vivo. Our data underscore the role of a perturbed, ineffective inflammatory response as a major contributor to the pathogenesis of DFUs, which is facilitated by FOXM1-mediated deregulation of recruitment of neutrophils and macrophages, revealing a potential therapeutic strategy.


Subject(s)
Diabetic Foot/genetics , Diabetic Foot/immunology , Forkhead Box Protein M1/immunology , Wound Healing/immunology , Adult , Aged , Animals , Cell Proliferation , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/immunology , Diabetic Foot/pathology , Disease Models, Animal , Female , Forkhead Box Protein M1/antagonists & inhibitors , Forkhead Box Protein M1/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Male , Mice, Inbred Strains , Middle Aged , Mouth Mucosa/physiology , Pyridines/pharmacology , Thiophenes/pharmacology , Transcriptome/physiology , Wound Healing/genetics
2.
Front Immunol ; 11: 498703, 2020.
Article in English | MEDLINE | ID: mdl-33633721

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease. It is characterized by the production of various pathogenic autoantibodies and is suggested to be triggered by increased type I interferon (IFN) signature. Previous studies have identified increased plasmablasts in the peripheral blood of SLE patients. The biological characteristics of SLE plasmablasts remain unknown, and few treatments that target SLE plasmablasts have been applied despite the unique cellular properties of plasmablasts compared with other B cell subsets and plasma cells. We conducted microarray analysis of naïve and memory B cells and plasmablasts (CD38+CD43+ B cells) that were freshly isolated from healthy controls and active SLE (n = 4, each) to clarify the unique biological properties of SLE plasmablasts. The results revealed that all B cell subsets of SLE expressed more type I IFN-stimulated genes. In addition, SLE plasmablasts upregulated the expression of cell cycle-related genes associated with higher FOXM1 and FOXM1-regulated gene expression levels than that in healthy controls. This suggests that a causative relationship exists between type I IFN priming and enhanced proliferative capacity through FOXM1. The effects of pretreatment of IFNα on B cell activation and FOXM1 inhibitor FDI-6 on B cell proliferation and survival were investigated. Pretreatment with IFNα promoted B cell activation after stimulation with anti-IgG/IgM antibody. Flow cytometry revealed that pretreatment with IFNα preferentially enhanced the Atk and p38 pathways after triggering B cell receptors. FDI-6 inhibited cell division and induced apoptosis in activated B cells. These effects were pronounced in activated B cells pretreated with interferon α. This study can provide better understanding of the pathogenic mechanism of interferon-stimulated genes on SLE B cells and an insight into the development of novel therapeutic strategies.


Subject(s)
B-Lymphocytes/immunology , Forkhead Box Protein M1/immunology , Gene Expression Regulation/drug effects , Immunologic Memory/drug effects , Interferon-alpha/pharmacology , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/drug effects , Adult , B-Lymphocytes/pathology , Female , Gene Expression Regulation/immunology , Humans , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/therapy , Male , Middle Aged
3.
Cancer Sci ; 110(12): 3650-3662, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31571332

ABSTRACT

Pediatric refractory solid tumors are aggressive malignant diseases, resulting in an extremely poor prognosis. KOC1, FOXM1, and KIF20A are cancer antigens that could be ideal targets for anticancer immunotherapy against pediatric refractory solid tumors with positive expression for these antigens. This nonrandomized, open-label, phase I clinical trial evaluated the safety and efficacy of the NCCV Cocktail-1 vaccine, which is a cocktail of cancer peptides derived from KOC1, FOXM1, and KIF20A, in patients with pediatric refractory solid tumors. Twelve patients with refractory pediatric solid tumors underwent NCCV Cocktail-1 vaccination weekly by intradermal injections. The primary endpoint was the safety of the NCCV Cocktail-1 vaccination, and the secondary endpoints were the immune response, as measured by interferon-r enzyme-linked immunospot assay, and the clinical outcomes including tumor response and progression-free survival. The NCCV Cocktail-1 vaccine was well tolerated. The clinical response of this trial showed that 4 patients had stable disease after 8 weeks and 2 patients maintained remission for >11 months. In 4, 8, and 5 patients, the NCCV Cocktail-1 vaccine induced the sufficient number of peptide-specific CTLs for KOC1, FOXM1, and KIF20A, respectively. Patients with high peptide-specific CTL frequencies for KOC1, FOXM1, and KIF20A had better progression-free survival than those with low frequencies. The findings of this clinical trial showed that the NCCV Cocktail-1 vaccine could be a novel therapeutic strategy, with adequate effects against pediatric refractory solid tumors. Future large-scale trials should evaluate the efficacy of the NCCV Cocktail-1 vaccination.


Subject(s)
Cancer Vaccines/immunology , Forkhead Box Protein M1/immunology , Kinesins/immunology , Neoplasms/therapy , RNA-Binding Proteins/immunology , Adolescent , Adult , Child , Female , Histocompatibility Antigens Class I/analysis , Humans , Male , Neoplasms/immunology , Neoplasms/mortality , Progression-Free Survival , T-Lymphocytes, Cytotoxic/immunology , Vaccination , Young Adult
4.
Int Immunopharmacol ; 68: 74-80, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30612087

ABSTRACT

Osteoarthritis (OA) is the most common inflammatory joint disease that is mainly characterized by articular cartilage destruction. Forkhead box M1 (FOXM1) is a transcription factor that acts as a critical mediator of inflammatory response. However, the role of FOXM1 in OA has not been investigated. Interleukin (IL)-1ß is a major proinflammatory cytokine, which is associated with cartilage destruction in the pathophysiology of OA. In the present study, we used IL-1ß to stimulate chondrocytes for the establishment of OA in vitro model. We found that FOXM1 was up-regulated in IL-1ß-induced chondrocytes. Knockdown of FOXM1 attenuated IL-1ß-caused decrease in cell viability. Knockdown of FOXM1 suppressed the IL-1ß-induced production of inflammatory cytokines including tumor necrosis factor (TNF)-α, and IL-6. Besides, several inflammatory mediators, such as nitric oxide (NO), prostaglandin E2 (PGE2), inducible nitric oxide synthases (iNOS), and cyclooxygenase-2 (COX-2) were also repressed by knockdown of FOXM1. FOXM1 silencing also inhibited the production of matrix metalloproteinases (MMPs) including MMP-3 and MMP-13. Furthermore, we found that knockdown of FOXM1 blocked the IL-1ß-induced NF-κB activation in chondrocytes. These findings indicated that FOXM1 might play an important role in the pathogenesis of OA, suggesting that FOXM1 might be a potential therapeutic target for the treatment of OA.


Subject(s)
Chondrocytes/immunology , Cytokines/immunology , Forkhead Box Protein M1/immunology , Osteoarthritis/immunology , Cells, Cultured , Forkhead Box Protein M1/genetics , Gene Silencing , Humans , NF-kappa B/immunology , RNA, Small Interfering/genetics
5.
Clin Exp Med ; 18(3): 413-420, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29804142

ABSTRACT

The aim of the present study was to investigate the expression of Forkhead box transcription M1 (FoxM1) and Forkhead box transcription P3 (FoxP3) in gastric cancer tissues in order to reveal any correlation between FoxM1, FoxP3 and clinicopathological parameters. Their clinical significance in gastric cancer was also investigated. Immunohistochemistry was used to detect the expression of FoxM1 and FoxP3 in gastric cancer and para-cancer tissues. The clinical significance of FoxM1 and FoxP3 in gastric cancer was explored, and the association between FoxM1 and FoxP3 was further analyzed. As a result, the overexpression of FoxM1 and FoxP3 was evident in gastric cancer (P < 0.001). FoxM1 overexpression was showed to be correlated with late AJCC stage (P = 0.025), while positive tumoral FoxP3 expression was associated with deeper invasion (P = 0.020), lymph node metastasis (P = 0.019) and later AJCC stage (P = 0.024). Overexpression of FoxM1 or FoxP3 was revealed to be negative prognostic factors for survival duration (P < 0.05), whereas only FoxM1 was shown to be independently associated with prognosisin gastric cancer after multivariate analysis (P = 0.020). A significant and positive correlation between FoxM1 and FoxP3 expressions was finally confirmed (P = 0.001). This significantly positive correlation between FoxM1 and FoxP3 prompts that FoxM1 may induce immune inhibition by recruiting FoxP3+ Tregs, leading to the progression of carcinogenesis, invasion and metastasis.


Subject(s)
Forkhead Box Protein M1/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Stomach Neoplasms/genetics , T-Lymphocytes, Regulatory/pathology , Adult , Aged , Female , Forkhead Box Protein M1/immunology , Forkhead Transcription Factors/immunology , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Stomach Neoplasms/diagnosis , Stomach Neoplasms/immunology , Stomach Neoplasms/mortality , Survival Analysis , T-Lymphocytes, Regulatory/immunology
6.
Biochem Biophys Res Commun ; 495(1): 1432-1439, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29158087

ABSTRACT

Functional abnormal airway epithelial cells, along with activated inflammatory cells, resulting in chronic airway inflammation, are considered as the characteristic of asthma. Fatty Acid Binding Protein 4 (FABP4) takes part in glucose and lipid homeostasis, and also have an important role in allergic airway inflammation. However, whether FABP4 influence barrier function of airway epithelial cells is unknown. In vivo, a HDM-induced murine model of asthma was obtained to assessed airway inflammation and protein expression of E-cadherin and Forkhead Box M1 (FoxM1). In vitro, 16-HBE was cultured and was treated with hrFABP4, siFABP4, FABPF4 inhibitor BMS, or FoxM1 inhibitor RCM-1. IL-4, IL-5, and IL-13 level was determined by ELISA. Transepithelial electrical resistance (TER), paracellular permeability and E-cadherin-special immunofluorescence were measured to value airway epithelial barrier function. Intracellular ROS production was determined by DCF-DA fluorescence. FABP4 inhibitor BMS alleviate airway inflammation and destruction of E-cad in allergic mouse. Treatment with HDM or hrFABP4 aggravated inflammatory response, damaged airway epithelial barrier, which could be inhibited by siFABP4 and BMS. Treatment with HDM or hrFABP4 also enhanced levels of FoxM1, and Inhibited FoxM1 suppressed HDM- and hrFABP4-induced inflammation and airway epithelial barrier dysfunction. In addition, H2O2 promoted FoxM1 expression, HDM and hrFABP4 induced-FoxM1 could be inhibited by NAC, leading to decreased inflammation and improved airway epithelial barrier. Upregulated ROS induced by FABP4 was of significance in activating FoxM1 leading to airway inflammation and epithelial barrier dysfunction.


Subject(s)
Alveolar Epithelial Cells/immunology , Asthma/immunology , Cell Membrane Permeability/immunology , Fatty Acid-Binding Proteins/immunology , Forkhead Box Protein M1/immunology , Reactive Oxygen Species/immunology , Respiratory Mucosa/immunology , Animals , Asthma/pathology , Male , Mice , Mice, Inbred BALB C , Respiratory Mucosa/pathology
7.
Sci Signal ; 10(475)2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28420752

ABSTRACT

Targeting a transcription factor, generally considered to be "nondruggable," may help to prevent asthma from developing.


Subject(s)
Asthma/immunology , Cytokines/immunology , Forkhead Box Protein M1/immunology , Lung/immunology , Pyroglyphidae/immunology , Animals , Asthma/drug therapy , Forkhead Box Protein M1/antagonists & inhibitors , Humans
8.
Sci Signal ; 10(475)2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28420758

ABSTRACT

Goblet cell metaplasia and excessive mucus secretion associated with asthma, cystic fibrosis, and chronic obstructive pulmonary disease contribute to morbidity and mortality worldwide. We performed a high-throughput screen to identify small molecules targeting a transcriptional network critical for the differentiation of goblet cells in response to allergens. We identified RCM-1, a nontoxic small molecule that inhibited goblet cell metaplasia and excessive mucus production in mice after exposure to allergens. RCM-1 blocked the nuclear localization and increased the proteasomal degradation of Forkhead box M1 (FOXM1), a transcription factor critical for the differentiation of goblet cells from airway progenitor cells. RCM-1 reduced airway resistance, increased lung compliance, and decreased proinflammatory cytokine production in mice exposed to the house dust mite and interleukin-13 (IL-13), which triggers goblet cell metaplasia. In cultured airway epithelial cells and in mice, RCM-1 reduced IL-13 and STAT6 (signal transducer and activator of transcription 6) signaling and prevented the expression of the STAT6 target genes Spdef and Foxa3, which are key transcriptional regulators of goblet cell differentiation. These results suggest that RCM-1 is an inhibitor of goblet cell metaplasia and IL-13 signaling, providing a new therapeutic candidate to treat patients with asthma and other chronic airway diseases.


Subject(s)
Allergens/toxicity , Forkhead Box Protein M1/antagonists & inhibitors , Goblet Cells/immunology , Interleukin-13/immunology , STAT6 Transcription Factor/immunology , Animals , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/immunology , Goblet Cells/pathology , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 3-gamma/immunology , Interleukin-13/genetics , Metaplasia/chemically induced , Metaplasia/genetics , Metaplasia/immunology , Metaplasia/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/immunology , STAT6 Transcription Factor/genetics
9.
Int J Oncol ; 50(5): 1655-1662, 2017 May.
Article in English | MEDLINE | ID: mdl-28393243

ABSTRACT

We performed a clinical trial using HLA-A24-binding peptide vaccines containing a combination of novel cancer-testis antigens and anti-angiogenic peptides for advanced gastric cancer (GC). Thirty-five GC patients who had shown resistance to the standard therapy were enrolled in this clinical trial using vaccinations with a mixture of multiple peptides derived from DEPDC1, URLC10, FoxM1, Kif20A and VEGFR1. The safety, the overall survival (OS), and the immunological responses based on an ELISPOT assay were determined to assess differences in patients who were HLA-A24-positive [24(+)] and HLA-A24-negative [24(-)]. No severe adverse effects were observed except for severe skin reactions in 4 patients. The differences in OS were not significant between patients who were 24(+) and 24(-). In the 24(+) group, patients who showed T cell responses specific to antigen peptides had a tendency towards better survival than those who showed no response, especially to the DEPDC1 peptide. The patients with local skin reactions had significantly better OS than the others. Peptide vaccine therapy was found to be safe and is expected to induce specific T cell responses in patients with advanced GC. The survival benefit of peptide vaccine monotherapy may not have been shown and further trials are needed to confirm these results.


Subject(s)
Cancer Vaccines/administration & dosage , HLA-A24 Antigen/immunology , Stomach Neoplasms/therapy , Vaccines, Subunit/administration & dosage , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/therapeutic use , Cancer Vaccines/immunology , Disease-Free Survival , Female , Forkhead Box Protein M1/immunology , Forkhead Box Protein M1/therapeutic use , GTPase-Activating Proteins/immunology , GTPase-Activating Proteins/therapeutic use , HLA-A24 Antigen/therapeutic use , Humans , Kaplan-Meier Estimate , Kinesins/immunology , Kinesins/therapeutic use , Male , Middle Aged , Neoplasm Proteins/immunology , Neoplasm Proteins/therapeutic use , Neoplasm Staging , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Treatment Outcome , Vaccines, Subunit/adverse effects , Vaccines, Subunit/immunology , Vascular Endothelial Growth Factor Receptor-1/immunology , Vascular Endothelial Growth Factor Receptor-1/therapeutic use
10.
Oncotarget ; 7(30): 48401-48411, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27351282

ABSTRACT

Application of dendritic cells (DCs) pulsed with tumor-associated antigens is considered attractive in immunotherapy for hepatocellular carcinoma (HCC). In order to efficiently prime tumor-associated antigens specific for cytotoxic T lymphocytes (CTLs), it is important that DCs present tumor-associated antigens on MHC class I. MHC class I generally present endogenous antigens expressed in the cytosol. In this study, we developed a new antigen delivery tool based on cross presentation of exogenous antigens in DCs by using cytoplasmic transduction peptide (CTP). CTP protein could transduce FoxM1 tumor antigen into the cytosol of DCs, and CTP-FoxM1 fusion protein could stimulate activation and maturation of DCs. DCs pulsed with CTP-FoxM1 could induce specific CTLs. More importantly, the immunity induced by DCs loaded with CTP-FoxM1 could significantly inhibit tumor growth and metastasis in HCC-bearing mice, which was more potent than that induced by DCs loaded with FoxM1 or CTP, alone. Our results indicate that DCs pulsed with CTP-FoxM1 might be a promising vaccine candidate for HCC therapy and provide new insight into the design of DC-based immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Carcinoma, Hepatocellular/therapy , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Liver Neoplasms/therapy , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/therapeutic use , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cytosol/immunology , Dendritic Cells/transplantation , Female , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/immunology , Forkhead Box Protein M1/therapeutic use , Genes, MHC Class I/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/therapeutic use , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL