Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Vet Comp Orthop Traumatol ; 37(1): 13-22, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37562428

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the feasibility of safe positioning of double 2.3-mm headless cannulated self-compression screws (HCS) in a small dog cadaveric sacroiliac luxation model and to compare the static rotational biomechanical properties of fixation repaired using two different screw systems with a minimally invasive osteosynthesis technique: double 2.3-mm HCS and a single 3.5-mm standard cortical screw placed in a lag fashion. STUDY DESIGN: A unilateral small dog sacroiliac luxation model was stabilized using double 2.3-mm HCS (n = 11) or a single 3.5-mm cortical screw (n = 11). Radiographic and computed tomography (CT) imaging analyses and biomechanical testing of rotational force on the sacroiliac joint of both fixations were performed. The maximum load at failure and failure modes of each fixation were recorded and compared. RESULTS: Fluoroscopically guided percutaneous application of double HCS was safe in a unilateral sacroiliac luxation model in small dogs without violation of the vertebral and ventral sacral foramen. Furthermore, resistance to rotational force applied on fixation of the sacroiliac joint repaired with double 2.3-mm HCS estimated by maximum failure load was significantly higher than that of a single 3.5-mm cortical screw (p < 0.001). CONCLUSION: Although this was an experimental cadaveric study, based on our results, the use of smaller double HCS may be beneficial as an alternative to the conventional single lag screw for stabilization of sacroiliac luxation in small dogs.


Subject(s)
Dog Diseases , Joint Dislocations , Humans , Dogs , Animals , Bone Screws/veterinary , Joint Dislocations/surgery , Joint Dislocations/veterinary , Fracture Fixation, Internal/veterinary , Sacroiliac Joint/surgery , Cadaver , Biomechanical Phenomena
2.
J Radiol Prot ; 41(3)2021 Aug 19.
Article in English | MEDLINE | ID: mdl-34030147

ABSTRACT

Large accelerator facilities for scientific research and particle accelerators for radiotherapy have been constructed in Korea. The environments requiring radiological protection procedures have changed substantially and the legal requirements have also increased. However, the same regulatory criteria that are applied to small radiation-generating devices also apply to large accelerator facilities. This study evaluates an approach that uses new criteria based on radiological risk to classify different types of large accelerator facilities. To inform this study, regulations and licensing procedures adopted in various countries were reviewed. The radiological risks from different types of particle accelerators and different types of accelerated particles were investigated in relation to current and planned large accelerators in Korea. The investigation was based on neutron yield and induced radioactivity calculations undertaken using the Monte-Carlo code FLUKA. Based on this analysis, we propose two categories for large accelerator facilities. In the process of the facility construction through to getting an operation permit, we also propose a graded approach, which would require changes in the Korean Nuclear Safety Act. A two-step licensing procedure with a prior review and a final permit for accelerator operation is very practical and conservative in the view of radiation protection. The review process for the revision of the Nuclear Safety Act is currently in progress and a major aspect of this revision reflects the results of this study.


Subject(s)
Particle Accelerators , Radiation Protection , Monte Carlo Method , Neutrons , Republic of Korea
3.
Int J Mol Med ; 46(6): 2185-2193, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33125115

ABSTRACT

The erythroid differentiation regulator 1 (Erdr1) protein has been studied for its role in various inflammatory skin diseases, including skin cancer, actinic keratosis and psoriasis. However, the therapeutic effects of Erdr1 on wound repair and its underlying mechanisms remain unknown. The present study aimed to investigate the effects of Erdr1 on wound healing in vitro and in vivo. The results demonstrated that treatment with recombinant Erdr1 enhanced wound healing in vivo and in vitro. In addition, Erdr1 increased the proliferation and migration of human dermal fibroblasts (HDFs). Notably, Erdr1 significantly induced the production of the chemoattractant C­C motif chemokine ligand 2 (CCL2) and recruited immune cells involved in wound healing. Treatment with recombinant Erdr1 induced the activation of the ERK1/1, p38 and JNK1/2 mitogen­activated protein (MAP) kinases. Treatment with specific inhibitors for MAP kinase inhibitors markedly suppressed cell proliferation and migration, and inhibited the production of CCL2 in HDFs. Furthermore, the inhibition of CCL2 with a neutralizing antibody significantly suppressed the recombinant Erdr1­induced proliferation and migration of HDFs. The wound healing activity of Erdr1 was comparable to that of epidermal growth factor. Taken together, these results demonstrated that Erdr1 promoted the proliferation and migration of HDFs and exhibited potent wound healing properties mediated by CCL2. Therefore, the results of the present study suggested that Erdr1 may be a potential therapeutic target for promoting wound healing.


Subject(s)
Chemokine CCL2/biosynthesis , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Wound Healing , Animals , Antibodies, Neutralizing/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Female , Humans , Mice, Inbred BALB C , Wound Healing/drug effects
4.
J Biol Chem ; 294(47): 17951-17961, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31597697

ABSTRACT

Naïve CD4+ T cells in the periphery differentiate into regulatory T cells (Tregs) in which Foxp3 is expressed for their suppressive function. NLRP3, a pro-inflammatory molecule, is known to be involved in inflammasome activation associated with several diseases. Recently, the expression of NLRP3 in CD4+ T cells, as well as in myeloid cells, has been described; however, a role of T cell-intrinsic NLRP3 in Treg differentiation remains unknown. Here, we report that NLRP3 impeded the expression of Foxp3 independent of inflammasome activation in Tregs. NLRP3-deficient mice elevate Treg generation in various organs in the de novo pathway. NLRP3 deficiency increased the amount and suppressive activity of Treg populations, whereas NLRP3 overexpression reduced Foxp3 expression and Treg abundance. Importantly, NLRP3 interacted with Kpna2 and translocated to the nucleus from the cytoplasm under Treg-polarizing conditions. Taken together, our results identify a novel role for NLRP3 as a new negative regulator of Treg differentiation, mediated via its interaction with Kpna2 for nuclear translocation.


Subject(s)
Cell Differentiation , Cell Nucleus/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , alpha Karyopherins/metabolism , Animals , Female , Forkhead Transcription Factors/metabolism , Inflammasomes/metabolism , Mice, Inbred C57BL , Models, Biological , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Protein Transport
5.
Front Immunol ; 10: 1269, 2019.
Article in English | MEDLINE | ID: mdl-31231389

ABSTRACT

Asthma is a common chronic inflammatory disease, which is characterized by airway hyperresponsiveness (AHR), high serum levels of immunoglobulin (Ig)E, and recruitment of various inflammatory cells such as eosinophils and lymphocytes. Korean traditional fermented foods have been reported to exert beneficial effects against allergic diseases such as asthma and atopic dermatitis. In this study, we investigated whether Staphylococcus succinus strain 14BME20 (14BME20) isolated from doenjang, a traditional high-salt-fermented soybean food of Korea, exerts suppressive effects on allergic airway inflammation in a murine model. Mice were orally administered with 14BME20, then sensitized and challenged with ovalbumin as an allergen. Administration of the 14BME20 significantly suppressed AHR and influx of inflammatory cells into the lungs and reduced serum IgE levels. Moreover, the proportion of T helper type 2 (Th2) cells and the production of Th2 cytokines were decreased in 14BME20-treated mice, whereas dendritic cells (DCs) with tolerogenic characteristics were increased. In contrast, oral administration of 14BME20 increased the proportion of CD4+CD25+Foxp3+ regulatory T (Treg) cells and the level of interleukin (IL)-10 in 14BME20-treated mice. Furthermore, 14BME20 induced maturation of tolerogenic DCs, and 14BME20-treated DCs increased Treg cell population in a co-culture system of DCs and CD4+ T cells. The addition of a neutralizing anti-IL-10 mAb to the culture of cells that had been treated with 14BME20 decreased the enhanced Treg cell population, thereby indicating that 14BME20-treated DCs increase Treg cell population via DC-derived IL-10. These results demonstrate that oral administration of 14BME20 suppresses airway inflammation by enhancing Treg responses and suggest that the 14BME20 isolated from doenjang may be a therapeutic agent for allergic asthma.


Subject(s)
Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Food Microbiology , Interleukin-10/metabolism , Staphylococcus/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Asthma/immunology , Asthma/metabolism , Asthma/pathology , Asthma/prevention & control , Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/prevention & control , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Disease Susceptibility , Female , Immune Tolerance , Immunoglobulin E/blood , Immunoglobulin E/immunology , Inflammation Mediators/metabolism , Mice
6.
Biochem Biophys Res Commun ; 514(3): 875-880, 2019 06 30.
Article in English | MEDLINE | ID: mdl-31084930

ABSTRACT

In addition to a role in translation, AIMP1 is secreted to affect various immune cells, such as macrophages, dendritic cells, B cells, and natural killer cells. However, the direct effects of AIMP1 on T cells have not yet been reported. In this study, we investigated whether AIMP1 could modulate T cell responses directly. Results revealed that AIMP1 significantly inhibited T cell receptor (TCR)-dependent activation and proliferation of CD4 T cells, as well as decreased TCR stimuli-induced Ca2+ influx in CD4 T cells. In addition, microscopic analysis revealed that lipid raft association in response to TCR engagement was significantly reduced in the presence of AIMP1, and the phosphorylation of PLCγ and PI3K was also down-regulated in CD4 T cells by AIMP1. Furthermore, AIMP1 specifically enhanced the differentiation of regulatory T (Treg) cells, while it had no effect on T helper type 1 (Th1), type 2 (Th2), and type 17 (Th17) cell differentiation. Collectively, these results indicate that AIMP1 affects T cells directly by down-regulating TCR signaling complex formation and inducing Treg cell differentiation in CD4 T cells.


Subject(s)
Cytokines/pharmacology , Lymphocyte Activation/drug effects , Membrane Microdomains/drug effects , Receptors, Antigen, T-Cell/genetics , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Calcium/immunology , Calcium/metabolism , Cell Differentiation/drug effects , Cytokines/genetics , Cytokines/immunology , Female , Gene Expression Regulation , Immunophenotyping , Ion Transport/drug effects , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/immunology , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Phosphorylation/drug effects , Primary Cell Culture , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/immunology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
7.
Int J Mol Sci ; 19(11)2018 Nov 19.
Article in English | MEDLINE | ID: mdl-30463229

ABSTRACT

Visfatin, a member of the adipokine family, plays an important role in many metabolic and stress responses. The mechanisms underlying the direct therapeutic effects of visfatin on wound healing have not been reported yet. In this study, we examined the effects of visfatin on wound healing in vitro and in vivo. Visfatin enhanced the proliferation and migration of human dermal fibroblasts (HDFs) and keratinocytes the expression of wound healing-related vascular endothelial growth factor (VEGF) in vitro and in vivo. Treatment of HDFs with visfatin induced activation of both extracellular signal-regulated kinases 1 and 2 (ERK1/2) and c-Jun N-terminal kinases 1 and 2 (JNK1/2) in a time-dependent manner. Inhibition of ERK1/2 and JNK1/2 led to a significant decrease in visfatin-induced proliferation and migration of HDFs. Importantly, blocking VEGF with its neutralizing antibodies suppressed the visfatin-induced proliferation and migration of HDFs and human keratinocytes, indicating that visfatin induces the proliferation and migration of HDFs and human keratinocytes via increased VEGF expression. Moreover, visfatin effectively improved wound repair in vivo, which was comparable to the wound healing activity of epidermal growth factor (EGF). Taken together, we demonstrate that visfatin promotes the proliferation and migration of HDFs and human keratinocytes by inducing VEGF expression and can be used as a potential novel therapeutic agent for wound healing.


Subject(s)
Cytokines/pharmacology , MAP Kinase Signaling System/drug effects , Nicotinamide Phosphoribosyltransferase/pharmacology , Wound Healing/drug effects , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Dermis/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Keratinocytes/metabolism , Mice, Inbred BALB C , Vascular Endothelial Growth Factor A/metabolism
8.
Front Immunol ; 9: 2095, 2018.
Article in English | MEDLINE | ID: mdl-30283443

ABSTRACT

T helper type 17 (Th17) cells are a subset of pro-inflammatory T helper cells that mediate host defense and pathological inflammation. We have previously reported that host dendritic cells (DCs) infected with Vibrio vulnificus induce Th17 responses through the production of several pro-inflammatory cytokines, including interleukin (IL)-1ß and IL-6. V. vulnificus produces RTX toxin (RtxA), an important virulence factor that determines successful pathophysiology. In this study, we investigated the involvement of RtxA from V. vulnificus in Th17 cell induction through the activation and maturation of DCs. The increased expression of the DC surface marker CD40 caused by V. vulnificus wild-type infection was reduced by rtxA gene mutation in V. vulnificus. The mRNA and protein levels of Th17 polarization-related cytokines also decreased in V. vulnificus rtxA mutant-infected DCs. In addition, the co-culture of Th cells and DCs infected with rtxA mutant V. vulnificus resulted in reduction in DC-mediated Th17 responses. Th17 cell responses in the small intestinal lamina propria decreased in mice inoculated with V. vulnificus rtxA mutant as compared to those inoculated with the wild-type strain. These decreases in DC maturation, Th17-polarizing cytokine secretion, and Th17 responses attributed to rtxA mutation were restored following infection with the rtxA revertant strain. Furthermore, the mutation in the hlyU gene encoding the activator of rtxA1 gene reproduced the results observed with rtxA mutation. Taken together, V. vulnificus, by means of RtxA, induces inflammatory Th17 responses, which may be associated with adaptive responses of the host against V. vulnificus infection.


Subject(s)
Bacterial Toxins/immunology , Inflammation/immunology , Th17 Cells/immunology , Vibrio Infections/immunology , Vibrio vulnificus/immunology , Animals , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Inflammation/metabolism , Inflammation/microbiology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Mice, Inbred C57BL , Th17 Cells/metabolism , Th17 Cells/microbiology , Vibrio Infections/metabolism , Vibrio Infections/microbiology , Vibrio vulnificus/physiology
9.
J Immunol ; 201(9): 2832-2841, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30275047

ABSTRACT

In addition to essential roles in protein synthesis, lysyl-tRNA synthetase (KRS) is secreted to trigger a proinflammatory function that induces macrophage activation and TNF-α secretion. KRS has been associated with autoimmune diseases such as polymyositis and dermatomyositis. In this study, we investigated the immunomodulatory effects of KRS on bone marrow-derived dendritic cells (DCs) of C57BL/6 mice and subsequent polarization of Th cells and analyzed the underlying mechanisms. KRS-treated DCs increased the expression of cell surface molecules and proinflammatory cytokines associated with DC maturation and activation. Especially, KRS treatment significantly increased production of IL-12, a Th1-polarizing cytokine, in DCs. KRS triggered the nuclear translocation of the NF-κB p65 subunit along with the degradation of IκB proteins and the phosphorylation of MAPKs in DCs. Additionally, JNK, p38, and ERK inhibitors markedly recovered the degradation of IκB proteins, suggesting the involvement of MAPKs as the upstream regulators of NF-κB in the KRS-induced DC maturation and activation. Importantly, KRS-treated DCs strongly increased the differentiation of Th1 cells when cocultured with CD4+ T cells. The addition of anti-IL-12-neutralizing Ab abolished the secretion of IFN-γ in the coculture, indicating that KRS induces Th1 cell response via DC-derived IL-12. Moreover, KRS enhanced the OVA-specific Th1 cell polarization in vivo following the adoptive transfer of OVA-pulsed DCs. Taken together, these results indicated that KRS effectively induced the maturation and activation of DCs through MAPKs/NF-κB-signaling pathways and favored DC-mediated Th1 cell response.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Lysine-tRNA Ligase/immunology , Th1 Cells/immunology , Animals , Dendritic Cells/cytology , Dendritic Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Lysine-tRNA Ligase/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , NF-kappa B/metabolism , Signal Transduction/immunology
10.
Int J Mol Med ; 41(1): 531-540, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115398

ABSTRACT

Vibrio vulnificus (V. vulnificus) is a gram-negative bacterium, which causes life-threatening septicemia and gastroenteritis through the consumption of contaminated seafood or wound infection. In addition, V. vulnificus infection is known to stimulate the production of several pro-inflammatory cytokines, which are associated with inflammatory responses mediated predominantly by dendritic cells (DCs), functioning as antigen-presenting cells. The present study aimed to investigate whether V. vulnificus infection induced the maturation and activation of murine DCs, which have the ability to polarize T helper (Th) cells into Th17 cells. Dysregulated Th17 cell responses are known to cause tissue damage, promoting the penetration of pathogens; however, Th17 cells are also involved in host defense against infection. Infection with V. vulnificus significantly increased the expression of cell surface molecules, including CD40, CD80 and major histocompatibility complex class II, leading to the maturation and activation of DCs. In the present study, the analysis of the cytokine profiles of DCs upon infection with V. vulnificus revealed the preferential production of interleukin-1ß (IL-1ß) and IL-6, through which V. vulnificus-infected DCs induced the polarization of Th17 cells when naïve CD4+ T cells were co-incubated. The reduction of Th17 cell generation through the use of anti-IL-6 neutralizing antibodies indicated that the Th17-polarizing capacity of V. vulnificus was predominantly dependent on DC-derived IL-6. The in vivo administration of V. vulnificus-infected DCs consistently increased the Th17 cell population in the lymph nodes of mice. Finally, the oral administration of V. vulnificus in mice also increased Th17 cell responses in the lamina propria of the small intestine. These results collectively demonstrated that V. vulnificus induced inflammatory Th17 cell responses via DCs, which may be associated with the immunopathological effects caused by V. vulnificus infection.


Subject(s)
Antigen-Presenting Cells/immunology , Dendritic Cells/immunology , Inflammation/immunology , Th17 Cells/immunology , Vibrio Infections/immunology , Animals , Antibodies, Neutralizing/immunology , B7-1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , Cell Polarity/immunology , Humans , Inflammation/genetics , Inflammation/microbiology , Inflammation/pathology , Interleukin-1beta/immunology , Interleukin-6/immunology , Lymph Nodes/immunology , Major Histocompatibility Complex/immunology , Mice , Vibrio Infections/genetics , Vibrio Infections/microbiology , Vibrio Infections/pathology , Vibrio vulnificus/immunology , Vibrio vulnificus/pathogenicity
11.
Int J Cancer ; 140(10): 2364-2374, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28224619

ABSTRACT

Treatment of acute myeloid leukemia (AML) largely depends on chemotherapy, but current regimens have been unsatisfactory for long-term remission. Although differentiation induction therapy utilizing 1,25(OH)2 D3 (VD3) has shown great promise for the improvement of AML treatment efficacy, severe side effects caused by its supraphysiological dose limit its clinical application. Here we investigated the combinatorial effect of l-asparaginase (ASNase)-mediated amino acid depletion and the latent alternation of VD3 activity on the induction of myeloid differentiation. ASNase treatment enhanced VD3-driven phenotypic and functional differentiation of three-different AML cell lines into monocyte/macrophages, along with c-Myc downregulation. Using gene silencing with shRNA and a chemical blocker, we found that reduced c-Myc is a critical factor for improving VD3 efficacy. c-Myc-dependent inhibition of mTORC1 signaling and induction of autophagy were involved in the enhanced AML cell differentiation. In addition, in a postculture of AML cells after each treatment, ASNase supports the antileukemic effect of VD3 by inhibiting cell growth and inducing apoptosis. Finally, we confirmed that the administration of ASNase significantly improved VD3 efficacy in the prolongation of survival time in mice bearing tumor xenograft. Our results are the first to demonstrate the extended application of ASNase, which is currently used for acute lymphoid leukemia, in VD3-mediated differentiation induction therapy for AML, and suggest that this drug combination may be a promising novel strategy for curing AML.


Subject(s)
Asparaginase/metabolism , Calcitriol/pharmacology , Cell Differentiation/drug effects , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Apoptosis/drug effects , Blotting, Western , Bone Density Conservation Agents/pharmacology , Cell Proliferation/drug effects , Down-Regulation , Female , Humans , Immunoenzyme Techniques , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Mol Med Rep ; 14(3): 2691-6, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27431807

ABSTRACT

Vibrio vulnificus is known to induce severely fulminant and fatal septicemia in susceptible hosts. In the present study, the antimicrobial activity of natural marine product-derived compounds against V. vulnificus, were investigated in vitro and in vivo. Twelve pure compounds were isolated from natural marine products and their inhibitory effects on V. vulnificus-induced cytotoxicity were determined in INT­407 cells. Among the 12 pure compounds tested, treatment with psammaplin A significantly suppressed V. vulnificus­induced cytotoxicity in INT­407 cells. Notably, treatment with psammaplin A (5-50 µg) had improved survival rates compared with that in the untreated mice, when the mice were infected with V. vulnificus intraperitoneally. In addition, the bacterial load of V. vulnificus in several tissues (spleen, liver and small intestine) was significantly lower in psammaplin A­treated mice than in untreated mice. Furthermore, psammaplin A treatment significantly suppressed the growth of V. vulnificus. Taken together, these results indicate that psammaplin A may be a potential agent for the prevention and treatment of V. vulnificus infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Aquatic Organisms/chemistry , Biological Products/pharmacology , Disulfides/pharmacology , Tyrosine/analogs & derivatives , Vibrio vulnificus/drug effects , Animals , Cell Line , Disease Models, Animal , Epithelial Cells , Female , Humans , Mice , Microbial Sensitivity Tests , Molecular Structure , Tyrosine/pharmacology , Vibrio Infections/drug therapy , Vibrio Infections/microbiology , Vibrio Infections/mortality , Vibrio Infections/pathology
13.
Cancer Immunol Immunother ; 65(1): 61-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26613952

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are one of the most important cell types that contribute to negative regulation of immune responses in the tumor microenvironment. Recently, aminoacyl-tRNA synthetase-interacting multifunctional protein 1 (AIMP1), a novel pleiotropic cytokine, was identified as an antitumor protein that inhibits angiogenesis and induces antitumor responses. However, the effect of AIMP1 on MDSCs in the tumor environment remains unclear. In the present study, we demonstrated that AIMP1 significantly inhibited tumor growth in 4T1 breast cancer-bearing mice and reduced MDSCs population of tumor sites and spleens of tumor-bearing mice. AIMP1 reduced expansion of MDSCs from bone marrow-derived cells in the tumor-conditioned media. AIMP1 also negatively regulated suppressive activities of MDSCs by inhibiting IL-6 and NO production, and Arg-1 expression. Furthermore, treatment of breast cancer-bearing mice with AIMP1 decreased the capacity of MDSCs to suppress T cell proliferation and Treg cell induction. Western blot and inhibition experiments showed that downregulation of MDSCs functions by AIMP1 may result from attenuated activation of STATs, Akt, and ERK. These findings indicate that AIMP1 plays an essential role in negative regulation of suppressive functions of MDSCs. Therefore, it has a significant potential as a therapeutic agent for cancer treatment.


Subject(s)
Amino Acyl-tRNA Synthetases/immunology , Antigen Presentation/immunology , Breast Neoplasms/immunology , Myeloid Cells/immunology , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...