Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Immunol Res ; 70(5): 708-713, 2022 10.
Article in English | MEDLINE | ID: mdl-35729473

ABSTRACT

AIM OF THE STUDY: This study aimed to analyze serum and cerebrospinal fluid (CSF) concentrations of proinflammatory and anti-inflammatory cytokines produced by T regulatory (Treg) cells in early RRMS according to the 2017 McDonald criteria. CLINICAL RATIONALE FOR THE STUDY: Multiple sclerosis (MS) is a chronic, inflammatory, demyelinating disease of the central nervous system (CNS) with the cytokine network playing an important role. However, there is a continual lack of data regarding the immunopathogenesis of early RRMS, especially according to the 2017 McDonald criteria. MATERIALS AND METHODS: The study groups included early RRMS patients during relapse (n = 18), remission (n = 14), and the control group. The MS diagnosis was established according to the 2017 McDonald criteria. Patients were studied up to 1 year after diagnosis was made. A quantitative test kit based on ELISA was used for cytokine measurement in the serum and CSF. Comparative and correlation analyses between the levels of TNF-α, TGF-ß2, IgG index, and relapse duration were performed. RESULTS: Significantly higher CSF concentrations of TNF-α in both RRMS-relapse and RRMS-remission groups were found compared to the controls (p < 0.01). The CSF levels of TGF-ß2 in the RRMS-relapse group were significantly lower in comparison to the control group (p = 0.01). CONCLUSIONS AND CLINICAL IMPLICATIONS: An inappropriate inflammatory response seems to occur in early RRMS and includes the production of TNF-α and a decrease in TGF-ß2 release suggesting a significant Treg cells role. Further studies on the topic may contribute to developing new disease-modifying drugs and biochemical markers of the disorder.


Subject(s)
Multiple Sclerosis, Relapsing-Remitting , Transforming Growth Factor beta2 , Tumor Necrosis Factor-alpha , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Cytokines , Humans , Immunoglobulin G , Multiple Sclerosis/blood , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/immunology , Recurrence , Transforming Growth Factor beta2/blood , Transforming Growth Factor beta2/cerebrospinal fluid , Transforming Growth Factor beta2/immunology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/cerebrospinal fluid , Tumor Necrosis Factor-alpha/immunology
2.
Front Immunol ; 12: 763877, 2021.
Article in English | MEDLINE | ID: mdl-35069536

ABSTRACT

Malignant melanoma is an aggressive form of cancer, which can be treated with anti-CTLA-4 and anti-PD-1 checkpoint inhibitor antibodies but while anti-CTLA-4 antibodies have clear benefits for some patients with melanoma, productive responses are difficult to predict and often associated with serious immune related adverse events. Antibodies specific to CTLA-4 bind two major isoforms of CTLA-4 in humans, the receptor isoform and a second naturally secretable, soluble isoform - sCTLA-4. The primary aim here was to examine the effect of selectively blocking the function of sCTLA-4 on in vitro immune responses from volunteer healthy or melanoma patient PBMC samples. Addition of recombinant sCTLA-4 to healthy PBMC samples demonstrated sCTLA-4 to have immunosuppressive capacity comparable to recombinant CTLA4-Ig, partially reversible upon antibody blockade. Further, we identified a mechanistic relationship where melanoma patient TGFß2 serum levels correlated with sCTLA-4 levels and provided the basis for a novel protocol to enhance sCTLA-4 production and secretion by T cells with TGFß2. Finally, a comparison of selective antibody blockade of sCTLA-4 demonstrated that both healthy and melanoma patient effector cytokine responses can be significantly increased. Overall, the data support the notion that sCTLA-4 is a contributory factor in cancer immune evasion.


Subject(s)
Antibodies, Neoplasm/immunology , CTLA-4 Antigen/immunology , Immune Checkpoint Inhibitors , Melanoma , Neoplasm Proteins/immunology , Transforming Growth Factor beta2/immunology , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Female , Humans , Male , Melanoma/immunology , Melanoma/therapy , Mice , Middle Aged
3.
Medicine (Baltimore) ; 99(46): e23024, 2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33181667

ABSTRACT

Transforming growth factor-beta (TGF-ß2) is an important cytokine regulating immune cell function. However, whether TGF-ß2 controls the invasion of colorectal cancer (CRC) by immune cells is unknown. Therefore, we evaluated the expression of TGF-ß2 using multiple databases and determined the relationship between TGF-ß2 expression and tumor immune infiltration defined by a set of genetic markers. The analysis demonstrated that the expression of TGF-ß2 is closely related to the outcome of many cancers, and this correlation was particularly strong in CRC. In addition, the increased expression of TGF-ß2 was significantly associated with the expression of various markers of specific immune cell subpopulations, and overexpression of TGF-ß2 was closely related to the prognosis of colon cancer patients. Moreover, TGF-ß2 was related to the prognosis and infiltration of the tumor by immune cells in CRC patients. The obtained results indicate that TGF-ß2 is a critical factor regulating the recruitment of immune cells and controls their infiltration into colorectal tumors. Thus, high expression of TGF-ß2 not only facilitates the prognosis in CRC patients, but also may provide a new target for the treatment of CRC.


Subject(s)
Colonic Neoplasms/mortality , Colorectal Neoplasms/mortality , Immunity, Cellular/genetics , Lymphocytes/immunology , Transforming Growth Factor beta2/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Humans , Kaplan-Meier Estimate , Prognosis
4.
J Leukoc Biol ; 108(5): 1673-1692, 2020 11.
Article in English | MEDLINE | ID: mdl-32794350

ABSTRACT

The success of using immune checkpoint inhibitors to treat cancers implies that inhibiting an immunosuppressive cytokine, such as TGF-ß2, could be a strategy to develop novel adjuvants for microbial vaccines. To develop nucleic acid based TGF-ß2 inhibitors, we designed three antisense oligonucleotides, designated as TIO1, TIO2, and TIO3, targeting the conserve regions identical in human and mouse TGF-ß2 mRNA 3'-untranslated region. In cultured immune cells, TIO3 and TIO1 significantly reduced the TGF-ß2 mRNA expression and protein production. In mice, the TIO3 and TIO1, when formulated in various microbial vaccines, significantly enhanced the antibody response to the vaccines, and the TIO3-adjuvanted influenza virus vaccine induced effective protection against the influenza virus challenge. In the immunized mice, TIO3 formulated in microbial vaccines dramatically reduced surface-bound TGF-ß2 expression on CD4+ T cells and CD19+ B cells in the lymph node (LN) cells and spleen cells; up-regulated the expression of CD40, CD80, CD86, and MHC II molecules on CD19+ B cells and CD11c+ dendritic cells; and promoted IFN-γ production in CD4+ T cells and CD8+ T cells in the LN cells. Overall, TIO3 or TIO1 could be used as a novel type of adjuvant for facilitating the microbial vaccines to elicit more vigorous and persistent antibody response by interfering with TGF-ß2 expression.


Subject(s)
Adjuvants, Immunologic/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Oligonucleotides/pharmacology , Transforming Growth Factor beta2/antagonists & inhibitors , Vaccination , Vaccines/pharmacology , Adjuvants, Immunologic/genetics , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Dogs , Humans , Madin Darby Canine Kidney Cells , Mice , Oligonucleotides/genetics , RAW 264.7 Cells , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/immunology , U937 Cells , Vaccines/genetics , Vaccines/immunology
5.
J Clin Invest ; 130(12): 6261-6277, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32817592

ABSTRACT

FOXP3+CD4+ regulatory T cells (Tregs) are critical for immune homeostasis and respond to local tissue cues, which control their stability and function. We explored here whether developmental endothelial locus-1 (DEL-1), which, like Tregs, increases during resolution of inflammation, promotes Treg responses. DEL-1 enhanced Treg numbers and function at barrier sites (oral and lung mucosa). The underlying mechanism was dissected using mice lacking DEL-1 or expressing a point mutant thereof, or mice with T cell-specific deletion of the transcription factor RUNX1, identified by RNA sequencing analysis of the DEL-1-induced Treg transcriptome. Specifically, through interaction with αvß3 integrin, DEL-1 promoted induction of RUNX1-dependent FOXP3 expression and conferred stability of FOXP3 expression upon Treg restimulation in the absence of exogenous TGF-ß1. Consistently, DEL-1 enhanced the demethylation of the Treg-specific demethylated region (TSDR) in the mouse Foxp3 gene and the suppressive function of sorted induced Tregs. Similarly, DEL-1 increased RUNX1 and FOXP3 expression in human conventional T cells, promoting their conversion into induced Tregs with increased TSDR demethylation, enhanced stability, and suppressive activity. We thus uncovered a DEL-1/αvß3/RUNX1 axis that promotes Treg responses at barrier sites and offers therapeutic options for modulating inflammatory/autoimmune disorders.


Subject(s)
Calcium-Binding Proteins/immunology , Cell Adhesion Molecules/immunology , Integrin beta3/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Calcium-Binding Proteins/genetics , Cell Adhesion Molecules/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Integrin beta3/genetics , Mice , Mice, Knockout , Signal Transduction/genetics , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/immunology
6.
Int Immunopharmacol ; 84: 106532, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32388013

ABSTRACT

Immune checkpoint blockade (ICB) has been a major breakthrough in various cancers including gastric cancer (GC), yet the clinical outcomes remain poor. Currently, epithelial-mesenchymal transition (EMT) has been reported to be associated with tumor mutational burden (TMB), which can cause lack of response to ICB. However, the underlying mechanism remains unknown. Members of the transforming growth factorß (TGFB) family are regarded as the main mediators of EMT, yet how TGFB2 drives EMT in GC is not fully understood. In this study, we found that overexpression of TGFB2 was correlated with poor prognosis in TGCA-STAD and four GEO GC datasets.Gene set enrichment analysis revealed that the EMT pathway was significantly enriched in the high TGFB2 expression group, whilst the TMB-related pathways including mismatch repair, base excision repair, and DNA replication were strongly enriched in the low expression group. Furthermore, EMT score analysis, WGCNA and functional analysis showed that TGFB2 was co-expressed with neurite-related pathways that might drive EMT. Also, CIBERSORT analysis revealed that tumor-infiltrating immune cells like T follicular helper cells might participate in the process of TGFB2 affecting TMB levels in GC. Moreover, in other various cancers, TGFB2 was also negatively correlated with TMB levels as well as ICB response. Overall, these results revealed that TGFB2 could play a vital role in linking EMT and TMB in GC, suggesting that TGFB2 may be a predictive therapeutic target for GC.


Subject(s)
Biomarkers, Tumor/immunology , Epithelial-Mesenchymal Transition , Mutation , Stomach Neoplasms/immunology , Transforming Growth Factor beta2/immunology , Aged , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality
7.
Nutrients ; 12(4)2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32326558

ABSTRACT

Human colostrum (HC) is a rich source of immune mediators that play a role in immune defences of a newly born infant. The mediators include transforming growth factor ß (TGF-ß) which exists in three isoforms that regulate cellular homeostasis and inflammation, can induce or suppress immune responses, limit T helper 1 cells (Th1) reactions and stimulate secretory immunoglobulin A (IgA) production. Human milk TGF-ß also decreases apoptosis of intestinal cells and suppresses macrophage cytokine expression. The aim of the study was to determine the concentration of TGF-ß2 in HC obtained from the mothers who delivered vaginally (VD) or by caesarean section (CS), and to compare the concentrations in HC from mothers who delivered at term (TB) or preterm (PB). In this study, 56% of preterm pregnancies were delivered via CS. The concentrations of TGF-ß2 were measured in HC from 299 women who delivered in the 1st Department of Obstetrics and Gynaecology, Medical University of Warsaw: 192 (VD), 107 (CS), 251 (TB), and 48 (PB). The colostrum samples were collected within 5 days post-partum. TGF-ß2 levels in HC were measured by the enzyme-linked immunosorbent assay (ELISA) test with the Quantikine ELISA Kit-Human TGF-ß2 (cat.no. SB250). Statistical significance between groups was calculated by the Student t-test using StatSoft Statistica 13 software. The mean TGF-ß2 concentration in patients who delivered at term or preterm were comparable. The levels of TGF-ß2 in HC were higher after preterm than term being 4648 vs. 3899 ng/mL (p = 0.1244). The delivery via CS was associated with higher HC concentrations of TGF-ß2. The levels of TGF-ß2 were significantly higher in HC after CS than VD (7429 vs. 5240 ng/mL; p = 0.0017). The data from this study suggest: caesarean section was associated with increased levels of TGF-ß2 in HC. The increased levels of TGF-ß2 in HC of women who delivered prematurely require further research. Early and exclusive breast-feeding by mothers after caesarean section and premature births with colostrum containing high TGF-ß2 levels may prevent the negative impact of pathogens which often colonize the gastrointestinal tract and may reduce the risk of chronic diseases in this group of patients.


Subject(s)
Cesarean Section , Colostrum/chemistry , Obstetric Labor, Premature/metabolism , Postpartum Period/metabolism , Transforming Growth Factor beta2/metabolism , Breast Feeding , Chronic Disease , Colostrum/immunology , Female , Gastroenteritis/microbiology , Gastroenteritis/prevention & control , Humans , Infant, Newborn , Pregnancy , Premature Birth/immunology , Prospective Studies , Risk , Transforming Growth Factor beta2/immunology , Transforming Growth Factor beta2/physiology
8.
J Reprod Immunol ; 137: 102857, 2020 02.
Article in English | MEDLINE | ID: mdl-31837543

ABSTRACT

Soluble isoforms of the non-classical Human Leukocyte Antigen (HLA)-G as well as Transforming Growth Factor (TGF)-ß is expressed in seminal plasma possibly influencing the pregnancy potential. We wanted to examine the association of seminal plasma sHLA-G, TGF-ß1, TGF-ß2 and TGFß3 with pregnancy success in a cohort of 127 couples and 4 single women attending fertility treatment with the use of assisted reproduction technologies (ART). Soluble HLA-G, TGF-ß1, TGF-ß2 and TGF-ß3 in seminal plasma did not fluctuate significantly over time. We did not find any impact of seminal plasma sHLA-G, TGF-ß1, TGF-ß2 and TGF-ß3 on time-to-pregnancy measured as number of treatment cycles. There was a significant association between concentrations of seminal plasma sHLA-G and HLA-G variations in the 3'untranslated region (3'UTR) of the HLA-G gene, supporting and extending previous findings. Furthermore, by comparing seminal plasma concentrations of sHLA-G, TGF-ß1, TGF-ß2 and TGF-ß3 in male subjects with reduced semen quality, male subjects with normal semen quality, and sperm donors, we found that TGF-ß2 was significantly lower, and TGF-ß3 was significantly higher, in seminal plasma from sperm donors. These findings suggest that TGF-ß isoforms may influence semen quality and fertility.


Subject(s)
HLA-G Antigens/metabolism , Infertility, Male/immunology , Semen/metabolism , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta3/metabolism , 3' Untranslated Regions/genetics , Adult , Cohort Studies , Female , HLA-G Antigens/analysis , HLA-G Antigens/genetics , HLA-G Antigens/immunology , Humans , Infertility, Male/therapy , Male , Middle Aged , Polymorphism, Genetic/immunology , Pregnancy , Promoter Regions, Genetic/genetics , Protein Isoforms/analysis , Protein Isoforms/immunology , Protein Isoforms/metabolism , Reproductive Techniques, Assisted , Semen/immunology , Semen Analysis , Tissue Donors , Transforming Growth Factor beta1/analysis , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/analysis , Transforming Growth Factor beta2/immunology , Transforming Growth Factor beta3/analysis , Transforming Growth Factor beta3/immunology , Young Adult
9.
Clin Exp Allergy ; 49(9): 1201-1213, 2019 09.
Article in English | MEDLINE | ID: mdl-31058363

ABSTRACT

BACKGROUND: Human milk (HM) transforming growth factor beta (TGF-ß) is critical for inflammation regulation and oral tolerance promotion. Previous reports suggested that variations in HM TGF-ß levels are associated with allergic outcomes. OBJECTIVE: We undertook a systematic review (PROSPERO 2017 CRD42017069920) to reassess the evidence on the relationships between HM TGF-ß and allergic outcomes in children. METHODS: Electronic bibliographic databases (MEDLINE, EMBASE and Cochrane Library) were systematically searched. Two independent reviewers screened reference lists, extracted the data and assessed risk of bias using the National Institute for Clinical Excellence methodological checklist. RESULTS: A total of 21 studies were identified. Sixteen studies assessed relationships between HM TGF-ß and risk of eczema; 14, allergic sensitization; nine, wheezing/asthma; six, food allergy; three, allergic rhinitis/conjunctivitis. Five cohorts (5/18, 28%) reported a protective effect of TGF-ß1, while 3 (3/10, 30%) suggested increased risk of allergic outcomes development and 1 (1/10, 10%), a protective effect of TGF-ß2 on eczema. Meta-analysis was not possible due to significant heterogeneity in methodology, age of outcome assessment and differing statistical approaches. 71% (15/21) of studies carried a high risk of bias. CONCLUSION AND CLINICAL RELEVANCE: In contrast with previous findings, we did not find strong evidence of associations between HM TGF-ß and allergic outcomes. Differences in studies' methodology and outcomes do not allow unconditional rejection or acceptance of the hypothesis that HM TGF-ß influences the risk of allergy development. Future studies on diverse populations employing standardized methods, accurate phenotyping of outcomes and evaluation of the effect of TGF-ß in combination with other HM immune markers, microbiome and oligosaccharides are required.


Subject(s)
Milk Hypersensitivity/immunology , Milk Proteins/immunology , Milk, Human/immunology , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta2/immunology , Female , Humans , Infant , Infant, Newborn , Male , Milk Hypersensitivity/pathology
10.
Front Immunol ; 9: 2222, 2018.
Article in English | MEDLINE | ID: mdl-30319659

ABSTRACT

Donor human milk (DHM) is submitted to Holder pasteurization (HoP) to ensure its microbiological safety in human milk banks but this treatment affects some of its bioactive compounds. The objective of this work was to compare the effects of HoP and high temperature short time (HTST) treatments on some bioactive compounds found in DHM. A total of 24 DHM batches were processed in a continuous HTST system (70, 72, and 75°C for 5-25 s) and by HoP (62.5°C for 30 min). The concentrations of immunoglobulins (Igs) A, G, and M, transforming growth factor-beta 2 (TGF-ß2), adiponectine, ghrelin, and leptin were measured using a multiplex system, whereas the concentration of epidermal growth factor (EGF) was determined by ELISA. In relation to Igs, IgG showed the highest preservation rates (87-101%) after HTST treatments, followed by IgA (54-88%) and IgM (25-73%). Ig retention after any of the HTST treatments was higher than after HoP (p < 0.001). Treatment times required to reduce the concentration of IgM by 90% (D-value) were 130, 88, and 49 s at 70, 72, and 75°C, while the number of degrees Celsius required to change the D-value by one factor of 10 (z-value) was 11.79°C. None of the heat treatments had a significant effect on the concentrations of TGF-ß2, EGF, adiponectin, and ghrelin. In contrast, leptin was detected only in 4 of the samples submitted to HoP, whereas it was present in all samples after the different HTST treatments, with retention rates ranging between 34 and 68%. Globally, the concentration of IgA, IgG, IgM, and leptin in DHM was significantly higher after HTST pasteurization performed in a continuous system designed to be used in human milk banks than after the HoP procedure that is routinely applied at present.


Subject(s)
Hot Temperature/adverse effects , Milk, Human/immunology , Pasteurization , Adiponectin/analysis , Adiponectin/chemistry , Adiponectin/immunology , Epidermal Growth Factor/analysis , Epidermal Growth Factor/immunology , Female , Ghrelin/analysis , Ghrelin/chemistry , Ghrelin/immunology , Humans , Immunoglobulins/analysis , Immunoglobulins/chemistry , Immunoglobulins/immunology , Leptin/analysis , Leptin/chemistry , Leptin/immunology , Milk Banks , Milk, Human/chemistry , Milk, Human/microbiology , Protein Denaturation , Time Factors , Tissue Donors , Transforming Growth Factor beta2/analysis , Transforming Growth Factor beta2/chemistry , Transforming Growth Factor beta2/immunology
11.
Front Immunol ; 9: 688, 2018.
Article in English | MEDLINE | ID: mdl-29686677

ABSTRACT

The promise of inducing immunological tolerance through regulatory T cell (Treg) control of effector T cell function is crucial for developing future therapeutic strategies to treat allograft rejection as well as inflammatory autoimmune diseases. In the current study, we used murine allograft rejection as a model to identify microRNA (miRNA) regulation of Treg differentiation from naïve CD4 cells. We performed miRNA expression array in CD4+ T cells in the draining lymph node (dLN) of mice which received syngeneic or allogeneic grafts to determine the molecular mechanisms that hinder the expansion of Tregs. We identified an increase in miRNA cluster 297-669 (C2MC) after allogeneic transplantation, in CD4+ T cells, such that 10 of the 27 upregulated miRNAs were all from this cluster, with one of its members, mmu-miR-466a-3p (miR-466a-3p), targeting transforming growth factor beta 2 (TGF-ß2), as identified through reporter luciferase assay. Transfection of miR-466a-3p in CD4+ T cells led to a decreased inducible FoxP3+ Treg generation while inhibiting miR-466a-3p expression through locked nucleic acid resulting in increased Tregs and a reduction in effector T cells. Furthermore, in vivo inhibition of miR-466a-3p in an allogeneic skin-graft model attenuated T cell response against the graft through an increase in TGF-ß2. TGF-ß2 was as effective as TGF-ß1 at both inducing Tregs and through adoptive transfer, mitigating host effector T cell response against the allograft. Together, the current study demonstrates for the first time a new role for miRNA-466a-3p and TGF-ß2 in the regulation of Treg differentiation and thus offers novel avenues to control inflammatory disorders.


Subject(s)
MicroRNAs/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta2/immunology , Animals , Cell Differentiation , Disease Models, Animal , Female , Forkhead Transcription Factors/immunology , Graft Rejection , Mice, Inbred C3H , Mice, Inbred C57BL , Transplantation, Homologous
12.
FEBS J ; 285(12): 2278-2291, 2018 06.
Article in English | MEDLINE | ID: mdl-29676849

ABSTRACT

The prognosis for patients with malignant glioma is very poor and thus the identification of new potential therapeutic targets is critically important. In this work, we report a previously unknown role for the homeobox transcription factor HOXC10 in regulating immunosuppressive gene expression in glioma cell lines and their proliferative and invasive capacities. Although HOXC10 expression is dysregulated in several types of tumors, its potential function in glioma was not known. We found that HOXC10 expression was upregulated in glioma compared with normal tissue, and that HOXC10 expression positively associated with high grading of glioma. In three independent datasets (REMBRANDT glioma, The Cancer Genome Atlas glioblastoma multiforme and GSE4412), HOXC10 upregulation was associated with short overall survival. In two glioma cell lines, HOXC10 knock-down inhibited cell proliferation, colony formation, migration and invasion, and promoted apoptosis. In addition, HOXC10 knock-down suppressed the expression of genes that are involved in tumor immunosuppression, including those for transforming growth factor-ß 2, PD-L2, CCL2 and TDO2. A ChIP assay showed that HOXC10 directly bound to the PD-L2 and TDO2 promoter regions. In summary, our results suggest that HOXC10 upregulation in glioma promotes an aggressive phenotype and induces immunosuppressive gene expression, supporting further investigation of the potential of HOXC10 as a therapeutic target in glioma.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Homeodomain Proteins/genetics , Programmed Cell Death 1 Ligand 2 Protein/genetics , Tryptophan Oxygenase/genetics , Apoptosis , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Databases, Genetic , Genes, Reporter , Glioblastoma/immunology , Glioblastoma/mortality , Glioblastoma/pathology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/immunology , Humans , Luciferases/genetics , Luciferases/metabolism , Neoplasm Grading , Neoplasm Invasiveness , Programmed Cell Death 1 Ligand 2 Protein/immunology , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Survival Analysis , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/immunology , Tryptophan Oxygenase/immunology
13.
Cell Mol Life Sci ; 74(23): 4329-4337, 2017 12.
Article in English | MEDLINE | ID: mdl-28913545

ABSTRACT

Proliferative vitreoretinal diseases such as diabetic retinopathy, proliferative vitreoretinopathy (PVR), and age-related macular degeneration are a leading cause of decreased vision and blindness in developed countries. In these diseases, retinal fibro(vascular) membrane (FVM) formation above and beneath the retina plays an important role. Gene expression profiling of human FVMs revealed significant upregulation of periostin. Subsequent analyses demonstrated increased periostin expression in the vitreous of patients with both proliferative diabetic retinopathy and PVR. Immunohistochemical analysis showed co-localization of periostin with α-SMA and M2 macrophage markers in FVMs. In vitro, periostin blockade inhibited migration and adhesion induced by PVR vitreous and transforming growth factor-ß2 (TGF-ß2). In vivo, a novel single-stranded RNAi agent targeting periostin showed the inhibitory effect on experimental retinal and choroidal FVM formation without affecting the viability of retinal cells. These results indicated that periostin is a pivotal molecule for FVM formation and a promising therapeutic target for these proliferative vitreoretinal diseases.


Subject(s)
Cell Adhesion Molecules/genetics , Choroidal Neovascularization/genetics , Diabetic Retinopathy/genetics , Macular Degeneration/genetics , Vitreoretinopathy, Proliferative/genetics , Actins/genetics , Actins/immunology , Animals , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/immunology , Choroidal Neovascularization/immunology , Choroidal Neovascularization/pathology , Choroidal Neovascularization/therapy , Diabetic Retinopathy/immunology , Diabetic Retinopathy/pathology , Diabetic Retinopathy/therapy , Gene Expression Regulation , Gene Silencing , Humans , Macular Degeneration/immunology , Macular Degeneration/pathology , Macular Degeneration/therapy , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Retina/immunology , Retina/pathology , Signal Transduction , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/immunology , Vitreoretinopathy, Proliferative/immunology , Vitreoretinopathy, Proliferative/pathology , Vitreoretinopathy, Proliferative/therapy , Vitreous Body/immunology , Vitreous Body/pathology
14.
J Pediatr Gastroenterol Nutr ; 65(3): e60-e67, 2017 09.
Article in English | MEDLINE | ID: mdl-28827481

ABSTRACT

BACKGROUND AND OBJECTIVES: Breast milk is a complex bioactive fluid that varies across numerous maternal and environmental conditions. Although breast-feeding is known to affect neonatal gut microbiome, the milk components responsible for this effect are not well-characterized. Given the wide range of immunological activity breast milk cytokines engage in, we investigated 3 essential breast milk cytokines and their association with early life gut microbiota. METHODS: A total of 52 maternal-child pairs were drawn from a racially diverse birth cohort based in Detroit, Michigan. Breast milk and neonatal stool specimens were collected at 1-month postpartum. Breast milk transforming growth factor (TGF)ß1, TGFß2, and IL-10 were assayed using enzyme-linked immunosorbent assays, whereas neonatal gut microbiome was profiled using 16S rRNA sequencing. RESULTS: Individually, immunomodulators TGFß1 and TGFß2 were significantly associated with neonatal gut microbial composition (R = 0.024, P = 0.041; R = 0.026, P = 0.012, respectively) and increased richness, evenness, and diversity, but IL-10 was not. The effects of TGFß1 and TGFß2, however, were not independent of one another, and the effect of TGFß2 was stronger than that of TGFß1. Higher levels of TGFß2 were associated with the increased relative abundance of several bacteria, including members of Streptococcaceae and Ruminococcaceae, and lower relative abundance of distinct Staphylococcaceae taxa. CONCLUSIONS: Breast milk TGFß concentration explains a portion of variability in gut bacterial microbiota composition among breast-fed neonates. Whether TGFß acts in isolation or jointly with other bioactive components to alter bacterial composition requires further investigation. These findings contribute to an increased understanding of how breast-feeding affects the gut microbiome-and potentially immune development-in early life.


Subject(s)
Breast Feeding , Gastrointestinal Microbiome , Interleukin-10/immunology , Milk, Human/immunology , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta2/immunology , Adult , Biomarkers/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant , Infant, Newborn , Interleukin-10/metabolism , Male , Middle Aged , Milk, Human/metabolism , Prospective Studies , Regression Analysis , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/metabolism
15.
Article in Japanese | MEDLINE | ID: mdl-27181235

ABSTRACT

Transforming growth factor (TGF)-ß family is a cytokine family with various biological processes and forms a highly homologous group of three mammalian isoforms, TGF-ß1, TGF-ß2, and TGF-ß3. Most of the attention on TGF-ß family in immunology has been mainly focused on TGF-ß1 in that TGF-ß1 induces anti-inflammatory regulatory T cells (Treg), and inflammatory T helper 17 (Th17) cells in combination with interleukin-6. Although little attention has been focused on the immunological roles of TGF-ß2 and TGF-ß3, the function of TGF-ß3 for maintaining immunological homeostasis has recently been identified such as the induction of Th17 cells and direct regulatory effects on humoral immunity. TGF-ß1 and TGF-ß3 shares similar anti-inflammatory or pro-inflammatory functions, but exhibits significantly different effects on fibrosis and chondrogenesis. For the clinical application of TGF-ßs, the mechanisms by which each TGF-ß isoform regulates immunity has to be elucidated. In this review, we provide an overview of the effects, cellular targets, and therapeutic potential of TGF-ßs on immune responses and autoimmune diseases.


Subject(s)
Autoimmune Diseases/immunology , Immunity, Humoral/immunology , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta2/immunology , Transforming Growth Factor beta3/immunology , Animals , Chondrogenesis , Fibrosis , Homeostasis/genetics , Homeostasis/immunology , Humans , Immunity, Humoral/genetics , Interleukin-6/physiology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
16.
Clin Exp Rheumatol ; 33(4 Suppl 92): S63-9, 2015.
Article in English | MEDLINE | ID: mdl-26457612

ABSTRACT

Transforming growth factor-betas (TGF-ßs) are multifunctional cytokines that have been implicated in the regulation of a broad range of biological processes, including cell proliferation, cell survival, and cell differentiation. The three isoforms identified in mammals, TGF-ß1, TGF-ß2, and TGF-ß3, have high sequence homology, bind to the same receptors, and show similar biological functions in many in vitro studies. However, analysis of the in vivo functions of the three isoforms and mice deficient for each cytokine reveals striking differences, illustrating their unique biological importance and functional non-redundancy. Although increasing evidence suggests that TGF-ß1 and, to a lesser extent, TGF-ß2 play an integral role in maintaining immune tolerance, the immunological role of TGF-ß3 has not been carefully evaluated. Recent studies have focused on the multifunctional role of TGF-ß3. In this review, we provide an overview of the role of TGF-ß3 in immunity, with comparison to TGF-ß1 and -ß2.


Subject(s)
Immune System/immunology , Inflammation Mediators/immunology , Inflammation/immunology , Signal Transduction , Transforming Growth Factor beta3/immunology , Animals , Humans , Immune System/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/prevention & control , Inflammation Mediators/metabolism , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/immunology , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism
17.
J Allergy Clin Immunol ; 135(6): 1476-85.e7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25725991

ABSTRACT

BACKGROUND: Chronic rhinosinusitis (CRS) with nasal polyps (NPs) in Western populations is associated with TH2 cytokine polarization. IL-25, an IL-17 family cytokine, was recently reported to induce TH2-type immune responses and to contribute to several allergic diseases, such as atopic dermatitis and asthma. However, the role of IL-25 in Asian patients with nasal polyposis remains unclear. OBJECTIVE: We sought to determine the role of IL-25 in Asian patients with nasal polyposis and CRS. METHODS: We investigated IL-25 expression and its cellular origins in NPs of human subjects using immunohistochemistry (IHC), quantitative RT-PCR, and ELISA of NP tissues. Correlations between IL-25 expression and expression of other inflammatory markers in NP tissues were also explored. Anti-IL-25 neutralizing antibody was administered in an ovalbumin- and staphylococcal enterotoxin B-induced murine NP model to confirm the function of IL-25 during nasal polypogenesis. RESULTS: IL-25 expression was upregulated in NP mucosa from patients with CRS with NPs compared with uncinate process tissue from control subjects and those with CRS without NPs. Overexpression of epithelial IL-25 was confirmed by using IHC, and double IHC staining showed that tryptase-positive cells were one of the main sources of IL-25 among immune cells. Furthermore, IL-17 receptor B levels were also increased in immune cells of patients with NPs compared with those in control subjects. In NPs IL-25 mRNA expression positively correlated with the expression of several inflammatory markers, including T-box transcription factor, RAR-related orphan receptor C, GATA3, eosinophil cationic protein, TGF-ß1, and TGF-ß2. IL-25 was more abundant in the murine NP model compared with control mice, and similar correlations between IL-25 and inflammatory markers were observed in murine models. Anti-IL-25 treatment reduced the number of polyps, mucosal edema thickness, collagen deposition, and infiltration of inflammatory cells, such as eosinophils and neutrophils. This treatment also inhibited expression of local inflammatory cytokines, such as IL-4 and IFN-γ. Furthermore, expression of CCL11, CXCL2, intercellular adhesion molecule 1, and vascular cell adhesion molecule 1 in the nasal mucosa was suppressed in the anti-IL-25-treated group. CONCLUSION: Our results suggest that IL-25 secreted from the sinonasal epithelia and infiltrating mast cells plays a crucial role in the pathogenesis of CRS with NPs in Asian patients. In addition, our results suggest the novel possibility of treating nasal polyposis with anti-IL-25 therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Gene Expression/drug effects , Interleukin-17/antagonists & inhibitors , Nasal Polyps/drug therapy , Rhinitis/drug therapy , Sinusitis/drug therapy , Adult , Animals , Case-Control Studies , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Chronic Disease , Disease Models, Animal , Eosinophil Cationic Protein/genetics , Eosinophil Cationic Protein/immunology , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/pathology , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , Gene Expression/immunology , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mice , Middle Aged , Nasal Polyps/complications , Nasal Polyps/genetics , Nasal Polyps/immunology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/immunology , Rhinitis/complications , Rhinitis/genetics , Rhinitis/immunology , Sinusitis/complications , Sinusitis/genetics , Sinusitis/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/immunology
18.
PLoS One ; 10(2): e0117608, 2015.
Article in English | MEDLINE | ID: mdl-25668313

ABSTRACT

Transforming growth factor (TGF)-ß2 is an important anti-inflammatory protein in milk and colostrum. TGF-ß2 supplementation appears to reduce gut inflammatory diseases in early life, such as necrotizing enterocolitis (NEC) in young mice. However, the molecular mechanisms by which TGF-ß2 protects immature intestinal epithelial cells (IECs) remain to be more clearly elucidated before interventions in infants can be considered. Porcine IECs PsIc1 were treated with TGF-ß2 and/or lipopolysaccharide (LPS), and changes in the cellular proteome were subsequently analyzed using two-dimensional gel electrophoresis-MS and LC-MS-based proteomics. TGF-ß2 alone induced the differential expression of 13 proteins and the majority of the identified proteins were associated with stress responses, TGF-ß and Toll-like receptor 4 signaling cascades. In particular, a series of heat shock proteins had similar differential trends as previously shown in the intestine of NEC-resistant preterm pigs and young mice. Furthermore, LC-MS-based proteomics and Western blot analyses revealed 20 differentially expressed proteins following treatment with TGF-ß2 in LPS-challenged IECs. Thirteen of these proteins were associated with stress response pathways, among which five proteins were altered by LPS and restored by TGF-ß2, whereas six were differentially expressed only by TGF-ß2 in LPS-challenged IECs. Based on previously reported biological functions, these patterns indicate the anti-stress and anti-inflammatory effects of TGF-ß2 in IECs. We conclude that TGF-ß2 of dietary or endogenous origin may regulate the IEC responses against LPS stimuli, thereby supporting cellular homeostasis and innate immunity in response to bacterial colonization, and the first enteral feeding in early life.


Subject(s)
Endotoxins/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Intestines/immunology , Stress, Physiological/physiology , Transforming Growth Factor beta2/metabolism , Animals , Cell Line , Heat-Shock Proteins/immunology , Heat-Shock Proteins/metabolism , Immunity, Innate/immunology , Lipopolysaccharides/immunology , Proteome/immunology , Proteome/metabolism , Signal Transduction/immunology , Stress, Physiological/immunology , Swine , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Transforming Growth Factor beta2/immunology
19.
Colloids Surf B Biointerfaces ; 113: 33-42, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24060928

ABSTRACT

Migration and epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) are main causes of central posterior capsule opacification after cataract extraction combined with intraocular lens (IOL) implantation. In this study, commercially available hydrophobic acrylic IOLs were first pretreated with atmospheric pressure glow discharge plasma to produce plenty of negatively charged chemical groups onto IOL surface, then polyethylenimine was deposited onto IOL surfaces as a precursor monolayer, and then anti-TGF-ß2 (anti-T) antibody and poly-l-lysine were sequentially deposited onto IOL surface for four cycles followed by another upmost monolayer of anti-T antibody via layer-by-layer self-assembly technique. After the fabrication of anti-T antibody multilayers on IOL surface, the surface characteristics of the anti-T antibody functionalized IOL, as well as its effect on LECs adhesion, proliferation, migration and EMT were then tested in this study. Our results revealed that anti-T antibody multilayers could be successfully immobilized onto IOL surfaces by plasma pretreatment and layer-by-layer self-assembly technique, and could keep stable for at least 3 months on IOL surface. The anti-T antibody immobilized in the multilayers on IOL surfaces showed good immunological activity by its specific antigen-antibody interaction with exogenous TGF-ß2. Anti-T antibody functionalized IOL surface was as smooth and flat as the untreated IOL surface. No difference in optical or physical properties was found between the anti-T antibody functionalized IOLs and the untreated IOLs. Compared with the untreated IOLs, the anti-T antibody functionalized IOL greatly inhibited LECs from migration and EMT, yet showed only transient inhibition to LECs adhesion and no inhibition to LECs proliferation. With these data, we demonstrate a simple, inexpensive, and feasible method to fabricate surface functionalized IOL for in situ capture and neutralization of TGF-ß2 in the capsular bag, which might be a possible solution to preventing posterior capsule opacification after cataract surgery.


Subject(s)
Antibodies/chemistry , Epithelial-Mesenchymal Transition/physiology , Lens, Crystalline/chemistry , Transforming Growth Factor beta2/immunology , Antibodies/immunology , Antibodies/pharmacology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Lenses, Intraocular , Photoelectron Spectroscopy , Surface Properties
20.
Immunobiology ; 218(9): 1200-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23643295

ABSTRACT

Macrophages treated with TGFß2 (TGFß2-Mϕ) and antigen are highly tolerogenic in vivo, and induce antigen-specific and long-lasting tolerance in both naïve and primed mice via induction of suppressor/regulatory T cells. In this study, we examined the molecular pathways, including the requirements for Smad-dependent signaling, that are involved in the induction and function of tolerogenic TGFß2-Mϕ. Treatment of murine macrophages with TGFß2 induced translocation of Smad2/3 to the nucleus, and impairment of Smad3-, but not Smad2-, dependent signaling inhibited the tolerogenic function of a TGFß2-treated murine macrophage cell line. Gene expression in murine macrophages treated with TGFß2 was evaluated by microarray analysis. The FcγRI gene was one of a number of immune-related genes differentially expressed in TGFß2-Mϕ, and appeared to be critical for tolerance in this system, since TGFß2-Mϕ from FcγRI deficient mice were unable to induce tolerance. The role that FcγRI plays in TGFß2-Mϕ-mediated tolerance is currently unclear. The results of this study provide important information about the factors that are critical for the induction of TGFß2-Mϕ-mediated tolerance, and a better understanding of these mechanisms could lead to the development of more effective tolerance-inducing strategies for the treatment of autoimmune/inflammatory diseases.


Subject(s)
Immune Tolerance , Macrophages/immunology , Receptors, IgG/metabolism , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta2/immunology , Active Transport, Cell Nucleus , Animals , Antigen Presentation , Cell Line , Cell Nucleus/metabolism , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , Microarray Analysis , Receptors, IgG/genetics , Signal Transduction/genetics , Smad2 Protein/metabolism , Smad3 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...