Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 269
Filter
1.
Exp Gerontol ; 164: 111831, 2022 07.
Article in English | MEDLINE | ID: mdl-35525396

ABSTRACT

AIMS: This study aimed to evaluate the impact of a 12-week calorie-restricted diet and recreational sports training on gene expressions IL-15, ATROGIN-1 and MURF-1 in skeletal muscle of T2D patients. METHODS: Older adults with T2D (n = 39, 60 ± 6.0 years, BMI 33.5 ± 0.6 kg/m2) were randomly allocated to Diet+Soccer (DS), Diet+Running (DR) or Diet (D). The training sessions were moderate-to-high-intensity and performed 3 × 40 min/week for 12-weeks. Gene expression from vastus lateralis muscle obtained by qRT-PCR, dual-energy X-ray and fasting blood testing measurements were performed before and after 12-weeks. Statistical analysis adopted were two-way ANOVA and Paired t-test for gene expression, and RM-ANOVA test for the remainder variables. RESULTS: Total body weight was reduced in ~4 kg representing body fat mass in all groups after 12-weeks (P < 0.05). HbA1c values decreased in all groups post-intervention. Lipids profile improved in the training groups (P < 0.05) after 12-weeks. ATROGIN-1 and MURF-1 mRNA reduced in the DS (1.084 ± 0.14 vs. 0.754 ± 1.14 and 1.175 ± 0.34 vs. 0.693 ± 0.12, respectively; P < 0.05), while IL-15 mRNA increased in the DR (1.056 ± 0.12 vs. 1.308 ± 0.13; P < 0.05) after 12-weeks intervention. CONCLUSION: Recreational training with a moderate calorie-restricted diet can downregulates the expression of atrophy-associated myokines and increases the expression of anti-inflammatory gene IL-15.


Subject(s)
Caloric Restriction , Diabetes Mellitus, Type 2 , Exercise , Muscle, Skeletal , Aged , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Exercise/physiology , Gene Expression , Humans , Interleukin-15/biosynthesis , Interleukin-15/genetics , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , SKP Cullin F-Box Protein Ligases/biosynthesis , SKP Cullin F-Box Protein Ligases/genetics , Tripartite Motif Proteins/biosynthesis , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
2.
Int J Mol Sci ; 22(19)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34638566

ABSTRACT

Natural killer (NK) cells, members of the innate immune system, play an important role in the rejection of HLA class I negative tumor cells. Hence, a therapeutic vaccine, which can activate NK cells in addition to cells of the adaptive immune system might induce a more comprehensive cellular response, which could lead to increased tumor elimination. Dendritic cells (DCs) are capable of activating and expanding NK cells, especially when the NFκB pathway is activated in the DCs thereby leading to the secretion of the cytokine IL-12. Another prominent NK cell activator is IL-15, which can be bound by the IL-15 receptor alpha-chain (IL-15Rα) to be transpresented to the NK cells. However, monocyte-derived DCs do neither secrete IL-15, nor express the IL-15Rα. Hence, we designed a chimeric protein consisting of IL-15 and the IL-15Rα. Upon mRNA electroporation, the fusion protein was detectable on the surface of the DCs, and increased the potential of NFκB-activated, IL-12-producing DC to activate NK cells in an autologous cell culture system with ex vivo-generated cells from healthy donors. These data show that a chimeric IL-15/IL-15Rα molecule can be expressed by monocyte-derived DCs, is trafficked to the cell surface, and is functional regarding the activation of NK cells. These data represent an initial proof-of-concept for an additional possibility of further improving cellular DC-based immunotherapies of cancer.


Subject(s)
Dendritic Cells/immunology , Interleukin-15/biosynthesis , Killer Cells, Natural/immunology , Receptors, Interleukin-15/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Dendritic Cells/drug effects , Electroporation , Humans , I-kappa B Kinase/biosynthesis , I-kappa B Kinase/genetics , Immunotherapy , Interleukin-15/chemistry , Interleukin-15/genetics , Killer Cells, Natural/drug effects , Leukocytes, Mononuclear , NF-kappa B/pharmacology , Primary Cell Culture , Receptors, Interleukin-15/chemistry , Receptors, Interleukin-15/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
3.
Front Immunol ; 12: 713304, 2021.
Article in English | MEDLINE | ID: mdl-34659203

ABSTRACT

Crosstalk between T cells, dendritic cells, and macrophages in temporal leukocyte clusters within barrier tissues provides a new concept for T cell activation in the skin. Activated T cells from these leukocyte clusters play critical roles in the efferent phase of allergic contact hypersensitivity (CHS). However, the cytokines driving maintenance and survival of pathogenic T cells during and following CHS remain mostly unknown. Upon epicutaneous allergen challenge, we here report that macrophages produce IL-27 which then induces IL-15 production from epidermal keratinocytes and dermal myeloid cells within leukocyte clusters. In agreement with the known role of IL-15 as a T cell survival factor and growth cytokine, this signaling axis enhances BCL2 and survival of skin T cells. Genetic depletion or pharmacological blockade of IL-27 in CHS mice leads to abrogated epidermal IL-15 production resulting in a decrease in BCL2 expression in T cells and a decline in dermal CD8+ T cells and T cell cluster numbers. These findings suggest that the IL-27 pathway is an important cytokine for regulating cutaneous T cell immunity.


Subject(s)
Hypersensitivity/immunology , Hypersensitivity/metabolism , Interleukin-15/biosynthesis , Interleukin-27/metabolism , Macrophages/immunology , Macrophages/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Allergens/immunology , Animals , Biomarkers , Disease Models, Animal , Disease Susceptibility , Humans , Hypersensitivity/pathology , Keratinocytes/immunology , Keratinocytes/metabolism , Mice , Myeloid Cells/immunology , Myeloid Cells/metabolism , Skin/immunology , Skin/metabolism , Skin/pathology , THP-1 Cells
4.
PLoS One ; 16(9): e0257185, 2021.
Article in English | MEDLINE | ID: mdl-34516566

ABSTRACT

PURPOSE: Tuberculosis (TB) is the leading cause of infectious disease related mortality, and only 10% of the infected individuals develop active disease. The likelihood of progression of latent tuberculosis infection (LTBI) to active TB disease is high in HIV infected individuals. Identification of HIV+ individuals at risk would allow treating targeted population, facilitating completion of therapy for LTBI and prevention of TB development. NK cells have an important role in T cell independent immunity against TB, but the exact role of NK cell subsets in LTBI and HIV is not well characterized. METHODS: In this study, we compared the expansion and function of memory like NK cells from HIV-LTBI+ individuals and treatment naïve HIV+LTBI+ patients in response to Mtb antigens ESAT-6 and CFP-10. RESULTS: In freshly isolated PBMCs, percentages of CD3-CD56+ NK cells were similar in HIV+LTBI+ patients and healthy HIV-LTBI+ individuals. However, percentages of CD3-CD56+CD16+ NK cells were higher in healthy HIV-LTBI+ individuals compared to HIV+LTBI+ patients. HIV infection also inhibited the expansion of memory like NK cells, production of IL-32α, IL-15 and IFN-γ in response to Mtb antigens in LTBI+ individuals. CONCLUSION: We studied phenotypic, functional subsets and activation of memory like-NK cells during HIV infection and LTBI. We observed that HIV+LTBI+ patients demonstrated suboptimal NK cell and monocyte interactions in response to Mtb, leading to reduced IL-15, IFN-γ and granzyme B and increased CCL5 production. Our study highlights the effect of HIV and LTBI on modulation of NK cell activity to understand their role in development of interventions to prevent progression to TB in high risk individuals.


Subject(s)
HIV Infections/complications , HIV Infections/immunology , Immunologic Memory , Killer Cells, Natural/immunology , Latent Tuberculosis/complications , Latent Tuberculosis/immunology , Adult , Cell Communication , Cell Proliferation , Chemokines/biosynthesis , Granzymes/biosynthesis , HIV Infections/pathology , Humans , Interferon-gamma/metabolism , Interleukin-15/biosynthesis , Interleukins/metabolism , Latent Tuberculosis/pathology , Lymphocyte Subsets/immunology , Monocytes/metabolism
5.
Protein Expr Purif ; 177: 105746, 2021 01.
Article in English | MEDLINE | ID: mdl-32916300

ABSTRACT

Periplasmic expression of recombinant proteins ensures the production of biologically active proteins in a correctly folded state with several key advantages. This research focused on the in-frame cloning of rhIL-15 in pET-20 (+) vector with pelB-leader sequence to direct the protein to the bacterial periplasm. The target construct periplasmic expression was evaluated in four strains, BL21 (DE3), BL21 (DE3) pLysS, Rosetta 2 (DE3) and Rosetta-gami 2 (DE3). Soluble periplasmic expression of IL-15 was highest in Rosetta-gami 2 (DE3) followed by Rossetta 2 (DE3) whereas negligible expression was observed with rest of two expression host. Best expression clone was selected for purification by dye ligand affinity chromatography. Purified rhIL-15 was characterized by SDS-PAGE, Western blotting and SEC-HPLC. This is the first report of functional recombinant human interleukin-15 being expressed and purified with yield of 120 mg/L in the periplasmic space of E. coli.


Subject(s)
Cloning, Molecular/methods , Interleukin-15/genetics , Periplasm/genetics , Animals , Cell Line , Cell Proliferation/drug effects , Chromatography, Affinity/methods , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Interleukin-15/biosynthesis , Interleukin-15/pharmacology , Mice , Periplasm/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Solubility , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
6.
J Biol Chem ; 295(28): 9596-9605, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32444497

ABSTRACT

Cyclic changes of the human endometrium, such as proliferation, secretion, and decidualization, occur during regular menstrual cycles. Heart- and neural crest derivatives-expressed transcript 2 (HAND2) is a key transcription factor in progestin-induced decidualization of human endometrial stromal cells (ESCs). It has been suggested that HAND2 regulates interleukin 15 (IL15), a key immune factor required for the activation and survival of uterine natural killer (uNK) cells. Activated uNK cells can promote spiral artery remodeling and secrete cytokines to induce immunotolerance. To date, no studies have evaluated the transcription factors that regulate IL15 expression in human ESCs. In the present study, we examined whether HAND2 controls IL15 transcriptional regulation in human ESCs. Quantitative RT-PCR and histological analyses revealed that HAND2 and IL15 levels increase considerably in the secretory phase of human endometrium tissues. Results from ChIP-quantitative PCR suggested that HAND2 binds to a putative HAND2 motif, which we identified in the upstream region of the human IL15 gene through in silico analysis. Using a luciferase reporter assay, we found that the upstream region of the human IL15 gene up-regulates reporter gene activities in response to estradiol and a progestin representative (medroxyprogesterone) in ESCs. The upstream region of the human IL15 gene also exhibited increasing responsiveness to transfection with a HAND2 expression vector. Of note, deletion and substitution variants of the putative HAND2 motif in the upstream region of IL15 did not respond to HAND2 transfection. These findings confirm that HAND2 directly up-regulates human IL15 transcription in ESCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Endometrium/metabolism , Interleukin-15/biosynthesis , Response Elements , Transcription, Genetic , Up-Regulation , Adult , Basic Helix-Loop-Helix Transcription Factors/genetics , Endometrium/cytology , Estradiol/pharmacology , Female , Humans , Interleukin-15/genetics , Middle Aged , Progestins/pharmacology , Stromal Cells/cytology , Stromal Cells/metabolism
7.
Cancer Immunol Res ; 8(3): 309-320, 2020 03.
Article in English | MEDLINE | ID: mdl-31953246

ABSTRACT

Hepatocellular carcinoma (HCC) is the fourth most common cause of cancer-related death in the world, and curative systemic therapies are lacking. Chimeric antigen receptor (CAR)-expressing T cells induce robust antitumor responses in patients with hematologic malignancies but have limited efficacy in patients with solid tumors, including HCC. IL15 and IL21 promote T-cell expansion, survival, and function and can improve the antitumor properties of T cells. We explored whether transgenic expression of IL15 and/or IL21 enhanced glypican-3-CAR (GPC3-CAR) T cells' antitumor properties against HCC. We previously optimized the costimulation in GPC3-CARs and selected a second-generation GPC3-CAR incorporating a 4-1BB costimulatory endodomain (GBBz) for development. Here, we generated constructs encoding IL15, IL21, or both with GBBz (15.GBBz, 21.GBBz, and 21.15.GBBz, respectively) and examined the ability of transduced T cells to kill, produce effector cytokines, and expand in an antigen-dependent manner. We performed gene-expression and phenotypic analyses of GPC3-CAR T cells and CRISPR-Cas9 knockout of the TCF7 gene. Finally, we measured GPC3-CAR T-cell antitumor activity in murine xenograft models of GPC3+ tumors. The increased proliferation of 21.15.GBBz T cells was at least in part dependent on the upregulation and maintenance of TCF-1 (encoded by TCF7) and associated with a higher percentage of stem cell memory and central memory populations after manufacturing. T cells expressing 21.15.GBBz had superior in vitro and in vivo expansion and persistence, and the most robust antitumor activity in vivo These results provided preclinical evidence to support the clinical evaluation of 21.15.GPC3-CAR T cells in patients with HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Glypicans/immunology , Immunotherapy, Adoptive/methods , Interleukin-15/immunology , Interleukins/immunology , Liver Neoplasms/therapy , Animals , Apoptosis/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation/physiology , Female , Glypicans/genetics , Humans , Interleukin-15/biosynthesis , Interleukin-15/genetics , Interleukins/biosynthesis , Interleukins/genetics , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Protein Expr Purif ; 148: 30-39, 2018 08.
Article in English | MEDLINE | ID: mdl-29596991

ABSTRACT

IL15, a member of the common γ chain receptor (γc) cytokine family, is gaining attention in recent years as one of the most promising anti-tumor agents. IL15 regulates T cell activation and proliferation, promotes the survival of CD8+ CD44hi memory T cells and is also essential for NK cell expansion and development. Despite the attraction of developing IL15 as an anti-cancer agent, production of recombinant IL15 has proven to be difficult due to the stringent control of IL15 expression at the transcriptional, translational and the post-translational levels. Furthermore, the bioactivity of IL15 fused to an extra functional domain that is isolated from mammalian cells is generally inferior to recombinant IL15 produced by E. coli. In this study, we report that Lysine 86 in IL15 is responsible for the instability in mammalian cells when its C-terminus is fused to the albumin binding scFv (IL15-A10m3). We demonstrate that K86A or K86R mutants increased the expression of the fusion protein from HEK293 cells. When the wild type IL15 is used for the fusion, no recombinant IL15 fusion was detected in the culture media. Additionally, we determined that the residue 112 in IL15 is critical for the bioactivity of IL15-A10m3. Examination of single and double mutants provides a better understanding of how IL15 engages with its receptor complex to achieve full signaling capacity. The results of our experiments were successfully applied to scale up production to levels up to 50 mg/L and >10 mg/L of >95% pure monomeric recombinant fusion proteins after a 2-step purification from culture media. More importantly, the recombinant fusion protein produced is fully active in stimulating T cell proliferation, when compared to the recombinant wild type IL15.


Subject(s)
Interleukin-15/genetics , Interleukin-15/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/genetics , Escherichia coli/genetics , HEK293 Cells , Humans , Hyaluronan Receptors/genetics , Interleukin-15/biosynthesis , Killer Cells, Natural/chemistry , Killer Cells, Natural/immunology , Recombinant Fusion Proteins/biosynthesis
9.
Scand J Med Sci Sports ; 28(1): 116-125, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28449327

ABSTRACT

In vitro and in vivo studies described the myokine IL-15 and its receptor IL-15Rα as anabolic/anti-atrophy agents, however, the protein expression of IL-15Rα has not been measured in human skeletal muscle and data regarding IL-15 expression remain inconclusive. The purpose of the study was to determine serum and skeletal muscle IL-15 and IL-15Rα responses to resistance exercise session and to analyze their association with myofibrillar protein synthesis (MPS). Fourteen participants performed a bilateral leg resistance exercise composed of four sets of leg press and four sets of knee extension at 75% 1RM to task failure. Muscle biopsies were obtained at rest, 0, 4 and 24 hours post-exercise and blood samples at rest, mid-exercise, 0, 0.3, 1, 2, 4 and 24 hours post-exercise. Serum IL-15 was increased by ~5.3-fold immediately post-exercise, while serum IL-15Rα decreased ~75% over 1 hour post-exercise (P<.001). Skeletal muscle IL-15Rα mRNA and protein expression were increased at 4 hours post-exercise by ~2-fold (P<.001) and ~1.3-fold above rest (P=.020), respectively. At 24 hours post-exercise, IL-15 (P=.003) and IL-15Rα mRNAs increased by ~2-fold (P=.002). Myofibrillar fractional synthetic rate between 0-4 hours was associated with IL-15Rα mRNA at rest (r=.662, P=.019), 4 hours (r=.612, P=.029), and 24 hours post-exercise (r=.627, P=.029). Finally, the muscle IL-15Rα protein up-regulation was related to Leg press 1RM (r=.688, P=.003) and total weight lifted (r=.628, P=.009). In conclusion, IL-15/IL-15Rα signaling pathway is activated in skeletal muscle in response to a session of resistance exercise.


Subject(s)
Interleukin-15/biosynthesis , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Receptors, Interleukin-15/biosynthesis , Resistance Training , Adult , Humans , Interleukin-15/blood , Protein Biosynthesis , Receptors, Interleukin-15/blood , Signal Transduction , Young Adult
10.
Peptides ; 99: 14-19, 2018 01.
Article in English | MEDLINE | ID: mdl-29097254

ABSTRACT

Recently we found that guanylin (Gn) and its receptor, guanylyl cyclase C (GC-C), are uniquely expressed in the mesenteric macrophages of some diet-resistant rats and that double-transgenic (dTg) rats overexpressing Gn and GC-C in macrophages demonstrate reduced fatty acid synthase and fat accumulation in fat tissue even when fed a high-fat diet (HFD). Lipid accumulation and fatty acid synthase mRNA levels in cocultured dTg rat adipocytes and macrophages were reduced compared with those in adipocytes cultured with WT rat macrophages. Here, we investigated whether Interleukin-15 (IL-15) derived from Gn-GC-C-expressing macrophages regulates lipid accumulation in adipocytes. IL-15 inhibited fatty acid synthase and lipid accumulation via STAT5 in cultured adipocytes. IL-15 mRNA and protein levels in the mesenteric fat of HFD-fed dTg rats were significantly higher than those of HFD-fed WT rats. Phosphorylated STAT5 levels in the mesenteric fat of HFD-fed dTg rats were increased compared with those of HFD-fed WT rats. In addition, the mRNA level of fatty acid synthase in the mesenteric fat was lower in HFD-fed dTg rats than in HFD-fed WT rats. These results support the hypothesis that IL-15 secreted from Gn-GC-C-expressing macrophages contributes to the inhibition of fatty acid synthase and lipid accumulation in adipocytes, leading to obesity resistance.


Subject(s)
Adipocytes/metabolism , Fatty Acid Synthase, Type I/metabolism , Gastrointestinal Hormones/biosynthesis , Interleukin-15/biosynthesis , Macrophages/metabolism , Natriuretic Peptides/biosynthesis , Receptors, Enterotoxin/biosynthesis , Adipocytes/cytology , Animals , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Fatty Acid Synthase, Type I/genetics , Gastrointestinal Hormones/genetics , Interleukin-15/genetics , Macrophages/cytology , Natriuretic Peptides/genetics , Rats , Rats, Transgenic , Receptors, Enterotoxin/genetics
11.
Sci Rep ; 7(1): 6028, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28729536

ABSTRACT

Diabetes is associated with impaired wound healing, which may be caused primarily by a deficiency in dendritic epidermal T cells (DETCs). In the epidermis, IL-15, IGF-1, and mTOR are known to regulate the maintenance of DETCs; however, it is unclear how these molecules may intersect to regulate DETC homeostasis in diabetes. Here, we show that the reduction of DETCs in the epidermis of diabetic mice is caused by altered homeostasis mediated by a reduction in IL-15 levels. Both impaired mTOR activation and reduction of IL-15 in the epidermis play important roles in DETC homeostasis. Moreover, IGF-1 drives keratinocytes to produce IL-15. The activation of IL-15 is dependent on mTOR, and conversely, mTOR regulates IGF-1 production in DETC, in a classic feedback regulatory loop. Our data suggest that in the setting of diabetes, reduced IGF-1, impaired mTOR pathway activation and reduced IL-15 in the epidermis function coordinately to promote altered DETC homeostasis and delayed skin wound closure.


Subject(s)
Epidermal Cells/metabolism , Homeostasis , Interleukin-15/biosynthesis , T-Lymphocyte Subsets/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Biomarkers , Diabetes Mellitus, Experimental , Epidermal Cells/immunology , Immunophenotyping , Insulin-Like Growth Factor I/metabolism , Keratinocytes/metabolism , Mice , Models, Biological , Signal Transduction , T-Lymphocyte Subsets/immunology , Wound Healing
12.
Appl Microbiol Biotechnol ; 101(13): 5267-5278, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28391504

ABSTRACT

It has been documented that the purification of inclusion bodies from Escherichia coli by size exclusion chromatography (SEC) may benefit subsequent refolding and recovery of recombinant proteins. However, loading volume and the high cost of the column limits its application in large-scale manufacturing of biopharmaceutical proteins. We report a novel process using polyethylene glycol (PEG) precipitation under denaturing conditions to replace SEC for rapid purification of inclusion bodies containing recombinant therapeutic proteins. Using recombinant human interleukin 15 (rhIL-15) as an example, inclusion bodies of rhIL-15 were solubilized in 7 M guanidine hydrochloride, and rhIL-15 was precipitated by the addition of PEG 6000. A final concentration of 5% (w/v) PEG 6000 was found to be optimal to precipitate target proteins and enhance recovery and purity. Compared to the previously reported S-200 size exclusion purification method, PEG precipitation was easier to scale up and achieved the same protein yields and quality of the product. PEG precipitation also reduced manufacturing time by about 50 and 95% of material costs. After refolding and further purification, the rhIL-15 product was highly pure and demonstrated a comparable bioactivity with a rhIL-15 reference standard. Our studies demonstrated that PEG precipitation of inclusion bodies under denaturing conditions holds significant potential as a manufacturing process for biopharmaceuticals from E. coli protein expression systems.


Subject(s)
Escherichia coli/genetics , Inclusion Bodies , Interleukin-15/biosynthesis , Interleukin-15/chemistry , Polyethylene Glycols/chemistry , Biopharmaceutics/methods , Chemical Precipitation , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Escherichia coli/chemistry , Escherichia coli/metabolism , Humans , Inclusion Bodies/chemistry , Interleukin-15/isolation & purification , Protein Denaturation , Protein Folding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/therapeutic use
13.
PLoS One ; 11(12): e0168404, 2016.
Article in English | MEDLINE | ID: mdl-27997584

ABSTRACT

Post-traumatic stress disorder is one of the common mental ailments that is triggered by exposure to traumatic events. Till date, the molecular factors conferring risk to the development of PTSD is not well understood. In this study, we have conducted a meta-analysis followed by hierarchical clustering and functional enrichment, to uncover the potential molecular networks and critical genes which play an important role in PTSD. Two datasets of expression profiles from Peripheral Blood Mononuclear Cells from 62 control samples and 63 PTSD samples were included in our study. In PTSD samples of GSE860 dataset, we identified 26 genes informative when compared with Post-deploy PTSD condition and 58 genes informative when compared with Pre-deploy and Post-deploy PTSD of GSE63878 dataset. We conducted the meta-analysis using Fisher, roP, Stouffer, AW, SR, PR and RP methods in MetaDE package. Results from the rOP method of MetaDE package showed that among these genes, the following showed significant changes including, OR2B6, SOX21, MOBP, IL15, PTPRK, PPBPP2 and SEC14L5. Gene ontology analysis revealed enrichment of these significant PTSD-related genes for cell proliferation, DNA damage and repair (p-value ≤ 0.05). Furthermore, interaction network analysis was performed on these 7 significant genes. This analysis revealed highly connected functional interaction networks with two candidate genes, IL15 and SEC14L5 highly enriched in networks. Overall, from these results, we concluded that these genes can be recommended as some of the potential targets for PTSD.


Subject(s)
Carrier Proteins/biosynthesis , Databases, Genetic , Gene Expression Regulation , Gene Regulatory Networks , Interleukin-15/biosynthesis , Leukocytes, Mononuclear/metabolism , Stress Disorders, Post-Traumatic/metabolism , Biomarkers/metabolism , Carrier Proteins/genetics , Humans , Interleukin-15/genetics , Stress Disorders, Post-Traumatic/genetics
14.
J Pathol ; 240(4): 425-436, 2016 12.
Article in English | MEDLINE | ID: mdl-27538697

ABSTRACT

Corpus-dominant lymphocytic gastritis (LyG) is characterized by CD8+ T-cell infiltration of the stomach epithelium by a so far uncharacterized mechanism. Although Helicobacter pylori is typically undetectable in LyG, patients respond to H. pylori antibiotic eradication therapy, suggesting a non-H. pylori microbial trigger for the disease. Comparative microbiota analysis of specimens from LyG, H. pylori gastritis and healthy controls precluded involvement of H. pylori in LyG but identified Propionibacterium acnes as a possible disease trigger. In addition, the natural killer group 2 member D (NKG2D) system and the proinflammatory cytokine interleukin (IL)-15 are significantly upregulated in the gastric mucosa of LyG patients, and gastric epithelial cells respond to microbe-derived stimuli, including live P. acnes and the microbial products short-chain fatty acids, with induction of NKG2D ligands. In contrast, H. pylori infection does not activate or even repress NKG2D ligands. Together, our findings identify P. acnes as a possible causative agent for LyG, which is dependent on the NKG2D system and IL-15 activation. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Gastritis/microbiology , Gram-Positive Bacterial Infections/immunology , Killer Cells, Natural/immunology , Lymphocytosis/microbiology , Propionibacterium acnes/isolation & purification , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy , Case-Control Studies , Cells, Cultured , Child , Female , Gastric Mucosa/immunology , Gastritis/immunology , Gastritis/pathology , Gram-Positive Bacterial Infections/pathology , Helicobacter pylori/immunology , Humans , Immunophenotyping , Inflammation Mediators/metabolism , Interleukin-15/biosynthesis , Interleukin-15/genetics , Ligands , Lymphocytosis/immunology , Male , Microbiota , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Propionibacterium acnes/immunology , RNA, Messenger/genetics , Stomach/immunology , Stomach/microbiology , Stomach/pathology , Up-Regulation , Young Adult
15.
Pathology ; 48(6): 602-7, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27567226

ABSTRACT

IL-15 is a key regulatory cytokine that shares many biological properties with IL-2. Recently, it has been shown that IL-15 could be up-regulated in T cell-mediated inflammatory disorders, such as rheumatoid arthritis and inflammatory bowel diseases. However, the role and expression of IL-15 in the inflammatory autoimmune disease Sjögren's syndrome (SS) has not been investigated. In the present study we evaluated the expression of IL-15 mRNA and protein in minor salivary gland (MSG) biopsy specimens and in human salivary gland epithelial cell (SGEC) cultures obtained from patients with primary SS (pSS) and compared their expression with that seen in normal healthy control subjects. IL-15 gene and protein analysis revealed that SGEC are able to produce IL-15. Results obtained demonstrated that the number of IL-15(+) cultured SGEC was significantly higher in cells derived from patients with pSS in comparison with SGEC from healthy subjects; similar results were obtained for IL-15 immunoreactivity by using immunohistochemistry that revealed a strong expression both in acinar and in ductal cells from pSS MSG. These studies could provide a rational basis to determine whether IL-15 could be a good candidate for anti-cytokine therapy in chronic inflammatory pSS diseases.


Subject(s)
Inflammation/immunology , Interleukin-15/immunology , Sjogren's Syndrome/immunology , Adult , Aged , Blotting, Western , Cells, Cultured , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Flow Cytometry , Humans , Immunohistochemistry , Interleukin-15/biosynthesis , Middle Aged , Real-Time Polymerase Chain Reaction , Salivary Glands/immunology , Salivary Glands/metabolism , Up-Regulation
16.
Oncotarget ; 7(14): 17579-90, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27007051

ABSTRACT

Professional antigen-presenting cells (APCs) are potent generators of tumor antigen-specific cytotoxic T lymphocytes (CTLs) for adoptive immunotherapy; however, generation of APCs is cumbersome, expensive, and subject to the tumor microenvironment. Artificial APCs (aAPCs) have been developed as a cost-effective alternative to APCs. We developed a cellular aAPC that efficiently generated alpha-fetoprotein (AFP)-specific CTLs. We genetically modified the human B cell lymphoma cell line BJAB with a lentiviral vector to establish an aAPC called BA15. The expression of AFP(158-166)-HLA-A*02:01 complex, CD80, CD86, and interleukin (IL)-15 in BA15 cells was assessed. The efficiency of BA15 at generating AFP-specific CTLs and the specific cytotoxicity of CTLs against AFP+ cells were also determined. BA15 cells expressed high levels of AFP(158-166) peptide, HLA-A2, CD80, CD86, and IL-15. BA15 cells also exhibited higher efficiency in generating AFP-specific CTLs than did dendritic cells. These CTLs had greater cytotoxicity against AFP+ hepatocellular carcinoma cells than did CTLs obtained from dendritic cells in vitro and in vivo. Our novel aAPC system could provide a robust platform for the generation of functional AFP-specific CTLs for adoptive immunotherapy of hepatocellular carcinoma.


Subject(s)
Antigen-Presenting Cells/immunology , Epitopes, T-Lymphocyte/immunology , Immunotherapy, Adoptive/methods , T-Lymphocytes, Cytotoxic/immunology , alpha-Fetoproteins/immunology , Animals , Antigen-Presenting Cells/metabolism , Antigens, Neoplasm/immunology , Cell Line, Tumor , HLA-A2 Antigen/immunology , Hep G2 Cells , Humans , Interleukin-15/biosynthesis , Interleukin-15/immunology , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Peptide Fragments/immunology , Random Allocation , Xenograft Model Antitumor Assays
17.
Nat Neurosci ; 19(2): 243-52, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26752157

ABSTRACT

Recovery from organ-specific autoimmune diseases largely relies on the mobilization of endogenous repair mechanisms and local factors that control them. Natural killer (NK) cells are swiftly mobilized to organs targeted by autoimmunity and typically undergo numerical contraction when inflammation wanes. We report the unexpected finding that NK cells are retained in the brain subventricular zone (SVZ) during the chronic phase of multiple sclerosis in humans and its animal model in mice. These NK cells were found preferentially in close proximity to SVZ neural stem cells (NSCs) that produce interleukin-15 and sustain functionally competent NK cells. Moreover, NK cells limited the reparative capacity of NSCs following brain inflammation. These findings reveal that reciprocal interactions between NSCs and NK cells regulate neurorepair.


Subject(s)
Brain/pathology , Encephalitis/pathology , Killer Cells, Natural/pathology , Neural Stem Cells/pathology , Aged , Aged, 80 and over , Animals , Brain Chemistry , Cell Proliferation , Cerebral Ventricles/pathology , Cytokines/biosynthesis , Encephalitis/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Immune Tolerance , Interleukin-15/biosynthesis , Interleukin-15/genetics , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/pathology , Neural Stem Cells/metabolism , Recovery of Function
18.
Int J Paediatr Dent ; 26(5): 351-6, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26440733

ABSTRACT

BACKGROUND: Coeliac disease is a chronic small intestinal immune-mediated enteropathy precipitated by exposure to dietary gluten, and possible relationships between coeliac disease and dental pathogenic conditions during childhood have been poorly investigated. AIM: The dental pulp plays a pivotal role in the immune defence against possible entry of pathogens from teeth, and the aim of this work was to investigate quantitative transcription levels of selected genes (IL-9, IL-11, IL-15, IL-18, IL-21, IL-27, MICA, IFN-γ) coding for pro-inflammatory immune innate activities in the pulp of primary teeth from healthy children and children with coeliac disease. DESIGN: The pulp from primary teeth of 10 healthy children and 10 children with coeliac disease was used to extract RNA and prepare cDNA for quantitative PCR transcription analysis employing commercial nucleotide probes for selected genes. RESULTS: In children with coeliac disease, the genes coding for pro-inflammatory cytokines IFN-γ, IL-11, IL-18, and IL-21 were significantly overexpressed, suggesting the possible importance of these cytokines in the relationships between coeliac disease and dental disorders. CONCLUSION: For the first time, we reported in dental pulp of children possible relationships between coeliac disease and modulation in transcription of cytokine-dependent inflammatory activities.


Subject(s)
Celiac Disease/complications , Cytokines/biosynthesis , Cytokines/genetics , Dental Pulp/immunology , Dental Pulp/metabolism , Inflammation/genetics , Inflammation/immunology , Child , Child, Preschool , Female , Gene Expression Regulation , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/genetics , Humans , Immunity, Innate , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-11/biosynthesis , Interleukin-11/genetics , Interleukin-15/biosynthesis , Interleukin-15/genetics , Interleukin-18/biosynthesis , Interleukin-18/genetics , Interleukin-9/biosynthesis , Interleukin-9/genetics , Interleukins/biosynthesis , Interleukins/genetics , Male , RNA/analysis , Real-Time Polymerase Chain Reaction , Tooth, Deciduous/immunology , Tooth, Deciduous/metabolism
19.
Cell Biol Int ; 40(2): 204-13, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26503216

ABSTRACT

Gastric cancer is the second leading cause of cancer-related mortality worldwide. Adoptive cell therapy (ACT) for gastric cancer is a novel therapy modality. However, the therapeutic effectiveness in vivo is still limited. The objective of this study was to assess the value of interleukin-15 (IL-15)-transferred cytokine-induced killer (CIK) cells in ACT for gastric cancer. IL-15-IRES-TK retroviral vector was constructed and transferred into the CIK cells. A gastric tumor-bearing nude mice model was constructed by subcutaneously injecting gastric cancer cells, BGC-823. Gastric tumor-bearing nude mice were randomly divided into three groups (five mice each group) and injected with physiological saline, CIK cells, and IL-15-IRES-TK-transfected CIK cells for 2 weeks, respectively. IL-15-IRES-TK-transferred CIK cells were prepared successfully and flow cytometry (FCM) analysis indicated that the transfection rate reached 85.7% after 5 days culture. In vivo experiment, we found that CIK cells retarded tumor growth by reducing tumor volume and tumor weight, as well as increasing tumor inhibition rate. Furthermore, IL-15-IRES-TK-transferred CIK cells showed a much stronger inhibition on tumor growth than CIK cells alone. Tumor morphology observation and growth indexes also showed that IL-15-transfected CIK cells had stronger cytotoxicity to tumor tissue than CIK cells. IL-15-IRES-TK transfection could elevate the effects of CIK cells to gastric carcinoma. The engineered CIK cells carrying IL-15-IRES-TK may be used in the ACT for gastric carcinoma, but prudent clinical trial is still indispensable.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-15/immunology , Natural Killer T-Cells/immunology , Stomach Neoplasms/therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/immunology , Cell Line, Tumor , Cytokine-Induced Killer Cells/drug effects , Humans , Interleukin-15/biosynthesis , Interleukin-15/genetics , Male , Mice , Mice, Nude , Random Allocation , Stomach Neoplasms/immunology , Xenograft Model Antitumor Assays
20.
J Immunol Methods ; 428: 50-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26678161

ABSTRACT

Interleukin-15 (IL-15) is a pleiotropic cytokine and a member of the four α-helix bundle family of cytokines which include IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21. IL-15 exhibits a broad biological activity and induces the differentiation and proliferation of T, B and natural killer (NK) cells. In this study, a DNA fragment containing the mature human IL-15 sequence was cloned into pPICZaA vector, generating a fusion protein with the alpha factor signal sequence in the N-terminus and 6×His as well as c-Myc tags in the C-terminus. The resulting plasmid was integrated into the genome of Pichia pastoris strain X-33. Recombinant yeast transformants with high-level recombinant human IL-15 (rhIL-15) production were identified, which secrete as much as 75 mg/L rhIL-15 after 3 days of induction by methanol. The rhIL-15 was purified by Ni(+)-NTA affinity chromatography, followed by DEAE anion exchange, yielding over 95% highly purified rhIL-15. Mass spectrometry and MALDI-TOF-TOF analysis showed the purified rhIL-15 had larger molecular weights than expected, due to different degrees of N-linked glycosylation. The biological activity of the rhIL-15 proteins was measured by its ability to enhance cellular proliferation of CTLL-2 and NK cells. The results demonstrate that the experimental procedure we have reported here can produce a large amount of active recombinant human IL-15 from P. pastoris.


Subject(s)
Interleukin-15/immunology , Interleukin-15/isolation & purification , Pichia/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Animals , Cell Line , Cell Proliferation , Humans , Interleukin-15/biosynthesis , Interleukin-15/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Recombinant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...