Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Mol Cancer ; 23(1): 45, 2024 02 29.
Article in English | MEDLINE | ID: mdl-38424542

ABSTRACT

BACKGROUND: In the myeloid compartment of the tumor microenvironment, CD244 signaling has been implicated in immunosuppressive phenotype of monocytes. However, the precise molecular mechanism and contribution of CD244 to tumor immunity in monocytes/macrophages remains elusive due to the co-existing lymphoid cells expressing CD244. METHODS: To directly assess the role of CD244 in tumor-associated macrophages, monocyte-lineage-specific CD244-deficient mice were generated using cre-lox recombination and challenged with B16F10 melanoma. The phenotype and function of tumor-infiltrating macrophages along with antigen-specific CD8 T cells were analyzed by flow cytometry and single cell RNA sequencing data analysis, and the molecular mechanism underlying anti-tumorigenic macrophage differentiation, antigen presentation, phagocytosis was investigated ex vivo. Finally, the clinical feasibility of CD244-negative monocytes as a therapeutic modality in melanoma was confirmed by adoptive transfer experiments. RESULTS: CD244fl/flLysMcre mice demonstrated a significant reduction in tumor volume (61% relative to that of the CD244fl/fl control group) 14 days after tumor implantation. Within tumor mass, CD244fl/flLysMcre mice also showed higher percentages of Ly6Clow macrophages, along with elevated gp100+IFN-γ+ CD8 T cells. Flow cytometry and RNA sequencing data demonstrated that ER stress resulted in increased CD244 expression on monocytes. This, in turn, impeded the generation of anti-tumorigenic Ly6Clow macrophages, phagocytosis and MHC-I antigen presentation by suppressing autophagy pathways. Combining anti-PD-L1 antibody with CD244-/- bone marrow-derived macrophages markedly improved tumor rejection compared to the anti-PD-L1 antibody alone or in combination with wild-type macrophages. Consistent with the murine data, transcriptome analysis of human melanoma tissue single-cell RNA-sequencing dataset revealed close association between CD244 and the inhibition of macrophage maturation and function. Furthermore, the presence of CD244-negative monocytes/macrophages significantly increased patient survival in primary and metastatic tumors. CONCLUSION: Our study highlights the novel role of CD244 on monocytes/macrophages in restraining anti-tumorigenic macrophage generation and tumor antigen-specific T cell response in melanoma. Importantly, our findings suggest that CD244-deficient macrophages could potentially be used as a therapeutic agent in combination with immune checkpoint inhibitors. Furthermore, CD244 expression in monocyte-lineage cells serve as a prognostic marker in cancer patients.


Subject(s)
Melanoma , Monocytes , Humans , Animals , Mice , Monocytes/metabolism , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Macrophages/metabolism , CD8-Positive T-Lymphocytes , Carcinogenesis/metabolism , Tumor Microenvironment , Signaling Lymphocytic Activation Molecule Family/metabolism
2.
Biomed Pharmacother ; 154: 113553, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35994815

ABSTRACT

To overcome the hurdles of immunotherapy, we investigated whether calcipotriol, a synthetic vitamin D analog, could overcome the immune evasion of glioblastoma multiforme (GBM) by modulating immune responses and the immunosuppressive tumor microenvironment. Administration of calcipotriol considerably reduced tumor growth. Both in vivo and in vitro studies revealed that CD8+T and natural killer (NK) cell gene signatures were enriched and activated, producing high levels of IFN-γ and granzyme B. In contrast, regulatory T cells (Treg) were significantly reduced in the calcipotriol-treated group. The expression of CD127, the receptor for thymic stromal lymphopoietin (TSLP), is elevated in CD4+T cells and potentially supports T-cell priming. Depleting CD4+T cells, but not NK or CD8+T cells, completely abrogated the antitumor efficacy of calcipotriol. These data highlight that the calcipotriol/TSLP/CD4+T axis can activate CD8+T and NK cells with a concomitant reduction in the number of Tregs in GBM. Therefore, calcipotriol can be a novel therapeutic modality to overcome the immune resistance of GBM by converting immunologically "cold" tumors into "hot" tumors. DATA AVAILABILITY: Data are available upon reasonable request. The RNA-seq dataset comparing the transcriptomes of control and calcipotriol-treated GL261 tumors is available from the corresponding author upon request.


Subject(s)
Glioblastoma , Vitamin D , CD8-Positive T-Lymphocytes , Calcitriol/analogs & derivatives , Glioblastoma/metabolism , Humans , Killer Cells, Natural , Lymphocyte Activation , Tumor Microenvironment , Vitamin D/metabolism
3.
Biochem Biophys Res Commun ; 363(2): 399-404, 2007 Nov 16.
Article in English | MEDLINE | ID: mdl-17869218

ABSTRACT

Heat shock protein 27 (HSP27) is an intracellular stress protein with the cytoprotective effect for a variety of noxious stresses. In this study, using a protein delivery system, we demonstrated the potential cytoprotective effect of HSP27 as a therapeutic protein in cardiac cells and ischemia/reperfusion animal model. We constructed a recombinant HSP27 fused to the protein transduction domain (PTD) derived from HIV-1 TAT protein. Purified recombinant TAT-HSP27 protein was efficiently delivered to H9c2 cells, and its transduction showed cytoprotective effect against the hypoxic stress. Moreover, transduction of TAT-HSP27 also attenuated hypoxia-induced apoptosis, which was accompanied by reduced caspase-3 activity. In addition, intraperitoneal injection of TAT-HSP27 into rat resulted in efficient protein transduction in heart tissues, decreased infarcted myocardium (control vs TAT-HSP27, 39.1% vs 29.5%, P<0.05) and preserved heart function (fractional shortening, 15.6% vs 33.4%, P<0.05), as determined at 7 d after I/R. These results suggest that the PTD-mediated delivery of HSP27 protein may represent a potential therapeutic strategy as protein drug for ischemic heart diseases.


Subject(s)
Genetic Therapy/methods , Heat-Shock Proteins/therapeutic use , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/therapy , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/prevention & control , Animals , Cardiotonic Agents , Heat-Shock Proteins/genetics , Male , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Transduction, Genetic , Transfection/methods , Treatment Outcome , Ultrasonography
4.
Aggress Behav ; 33(1): 26-37, 2007.
Article in English | MEDLINE | ID: mdl-17441003

ABSTRACT

It has been proven extremely difficult in the past to estimate the prevalence of physical aggression in children for two main reasons: (a) a heterogeneous sampling of behaviors (i.e., mix between physically aggressive and non-physically aggressive antisocial behaviors), and (b) a lack of a "gold standard" to identify children who exhibit physically aggressive behaviors on a frequent basis. The goal of this study was to test for age differences in the prevalence of physical aggression in the Canadian population of school-aged boys and girls, using cross-sectional data from the National Longitudinal Survey of Children and Youth (NLSCY). The first wave of the NLSCY included a representative sample of 12,292 Canadian children aged 5-11 years. We used latent class analysis to identify children whose propensity to exhibit physically aggressive behaviors was much higher than that of other children of the same age and sex in the population. The prevalence of physical aggression was estimated at 3.7% in 5-11-year-old boys and ranged from .5% to 2.3% in 11 and 5-year-old girls, respectively. Hence, the results show a decreasing trend in the prevalence of physical aggression with age for girls, but not for boys. These findings suggest the importance of considering the developmental pathways of physical aggression for boys and girls separately.


Subject(s)
Aggression , Age Factors , Aggression/physiology , Canada/epidemiology , Child , Child, Preschool , Cross-Sectional Studies , Female , Humans , Male , Prevalence , Sex Characteristics
5.
Mol Cells ; 20(3): 429-34, 2005 Dec 31.
Article in English | MEDLINE | ID: mdl-16404160

ABSTRACT

Lysophosphatidylcholine (lysoPC) induces vascular smooth muscle cell (VSMC) proliferation and migration, which has been proposed to initiate the intimal thickening in coronary atherosclerotic lesions. Berberine is an alkaloid in Berberis aquifolium and many other plants. Recently, it has been shown to have beneficial effects on the cardiovascular system, such as anti-hyperglycemic and cholesterol-lowering activity. In this study, we investigated its effects on lysoPC-induced VSMC proliferation and migration. Berberine inhibited lysoPC-induced DNA synthesis and cell proliferation in VSMCs, as well as migration of the lysoPC-stimulated VSMCs. It also inhibited the activation of extracellular signal-regulated kinases (ERKs) and reduced transcription factor AP-1 activity and the lysoPC-induced increases in intracellular reactive oxygen species (ROS). These results indicate that the inhibitory effects of berberine on lysoPC-stimulated VSMC proliferation and migration are attributable to inhibition of ROS generation and hence of activation of the ERK1/2 pathway. This suggests that berberine has potential in the prevention of atherosclerosis and restenosis.


Subject(s)
Berberine/pharmacology , Lysophosphatidylcholines/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Thymidine/metabolism , Transcription Factor AP-1/metabolism
6.
Eur J Pharmacol ; 484(2-3): 119-25, 2004 Jan 26.
Article in English | MEDLINE | ID: mdl-14744595

ABSTRACT

Reactive oxygen species-mediated cellular injury is involved in the pathogenesis of many diseases, including those affecting the cardiovascular system, such as myocardial ischemia-reperfusion injury, inflammation, and atheroscleosis. Raxofelast (IRFI-016; (+/-)-5-acetoxy-2, 3-dihydro-4, 6, 7-trimethyl-2-benzofuran-acetic acid) was designed with the aim of maximizing the antioxidant potency of phenols chemically related to vitamin E. The antioxidant activity of raxofelast has been convincingly demonstrated in several in vitro studies and in various models of ischemia-reperfusion injury. In this study, the antiproliferative effects of raxofelast were investigated to determine whether transduction signals and protooncogenes are affected in H(2)O(2)-stimulated rat aortic smooth muscle cells. In a tetrazolium-based colorimetric assay, the proliferation of rat aortic smooth muscle cells was increased by 3-fold in 0.1% fetal bovine serum/Dulbecco's modified Eagle's medium (DMEM) containing 500 microM H(2)O(2), indicating that exogenous 500 microM H(2)O(2) was a growth stimulator of rat aortic smooth muscle cells. Exogenous H(2)O(2) significantly activated extracellular signal-regulated kinases (ERKs) activity within 30 min and raxofelast inhibited the ERKs activation dose dependently in 500 microM H(2)O(2)-stimulated rat aortic smooth muscle cells (IC(50): 200 microM). Raxofelast reduced the intracellular reactive oxygen species generated by exogenous H(2)O(2) in a dose-dependent manner. In 500 microM H(2)O(2)-stimulated rat aortic smooth muscle cells, raxofelast dramatically attenuated the activation of mitogen-activating protein kinase (MAPK)/ERK kinase 1, 2 (MEK1,2) and protein kinase C (PKC) without affecting Ras expression. Induction of c-myc mRNA was significantly reduced dose dependently up to 100 microM by raxofelast in concentrations. These data indicate that the antiproliferative effects of raxofelast in H(2)O(2)-stimulated rat aortic smooth muscle cells may involve the suppression of intracellular reactive oxygen species formation and the inhibition of ERKs by inactivation through PKC and MEK1,2 and down-regulation of c-myc expression, regardless of Ras activation.


Subject(s)
Aorta, Thoracic/drug effects , Benzofurans/pharmacology , Growth Inhibitors/pharmacology , Hydrogen Peroxide/pharmacology , Muscle, Smooth, Vascular/drug effects , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/physiology , Cell Division/drug effects , Cell Division/physiology , Dose-Response Relationship, Drug , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Sprague-Dawley
7.
Biochem Biophys Res Commun ; 305(4): 789-96, 2003 Jun 13.
Article in English | MEDLINE | ID: mdl-12767899

ABSTRACT

TNF-related weak inducer of apoptosis (TWEAK) is a member of the TNF ligand family that induces angiogenesis in vivo. The TWEAK receptor (TweakR) is a recently identified member of the TNF receptor (TNFR) superfamily and is expressed on smooth muscle cells (SMCs) and endothelial cells (ECs). In this report we identify the TNF receptor-associated factor (TRAF) family of signal transducers as important components of TweakR-mediated NF-kappa B activation. Coimmunoprecipitation experiments suggested potential interactions between the cytoplasmic tail of TweakR with TRAFs 1, 2, 3, and 5. Dominant negative forms of TRAF2 and TRAF5 substantially inhibited TweakR-mediated NF-kappa B activation, suggesting a role of TRAFs in regulating smooth muscle and endothelial cell function. Using alanine-scanning analysis, we defined a TRAF-binding motif, PIEET, in TweakR that mediates TRAF binding and NF-kappa B activation. Furthermore, TweakR mutations within the TRAF-binding motif abolished TweakR-stimulated SMC migration, revealing a role for TRAFs in TweakR-induced activation events.


Subject(s)
Muscle, Smooth, Vascular/metabolism , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Amino Acid Motifs , Animals , Binding Sites , Cell Line , Cell Movement , Muscle, Smooth, Vascular/physiology , Mutation , Proteins/genetics , Proteins/metabolism , Rats , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/classification , Signal Transduction , TNF Receptor-Associated Factor 2 , TNF Receptor-Associated Factor 5 , TWEAK Receptor , Transcriptional Activation
8.
J Steroid Biochem Mol Biol ; 84(1): 101-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12648529

ABSTRACT

In cardiac myocytes, stimulation of alpha(1)-adrenoceptor (AR) leads to a hypertrophic phenotype. The G(h) protein (transglutaminase II, TGII) is tissue type transglutaminase and transmits the alpha(1B)-adrenoceptor signal with GTPase activity. Recently, it has been shown that the calreticulin (CRT) down-regulates both GTP binding and transglutaminase activities of TGII. To elucidate whether G(h) mediates norepinephrine-stimulated intracellular signal transductions leading to activation of extracellular signal-regulated kinases (ERKs) and neonatal rat cardiomyocyte hypertrophy, we examined the effects of G(h) on the activation of ERKs and inhibitory effects of CRT on alpha(1)-adrenoceptor/G(h) signaling. In neonatal rat cardiomyocytes, norepinephrine-induced ERKs activation was inhibited by an alpha(1)-adrenoceptor blocker (prazosin), but not by an beta-adrenoceptor blocker (propranolol). Overexpression of the G(h) protein stimulated norepinephrine-induced ERKs activation, which was inhibited by alpha-adrenoceptor blocker (prazosin). Co-overexpression of G(h) and CRT abolished norepinephrine-induced ERKs activation. Taken together, norepinephrine induces hypertrophy in neonatal rat cardiomyocytes through alpha(1)-AR stimulation and G(h) is partly involved in norepinephrine-induced MEK1,2/ERKs activation. Activation of G(h)-mediated MEK1,2/ERKs was completely inhibited by CRT.


Subject(s)
Calreticulin/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Adrenergic alpha-Agonists/pharmacology , Animals , Animals, Newborn , Calreticulin/metabolism , Cells, Cultured , Down-Regulation , Enzyme Activation , Immunoblotting , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mitogen-Activated Protein Kinase 3 , Myocardium/cytology , Norepinephrine/pharmacology , Protein Binding , Rats , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction , Time Factors , Transfection
9.
Taehan Kanho Hakhoe Chi ; 33(2): 189-99, 2003 Apr.
Article in Korean | MEDLINE | ID: mdl-15314447

ABSTRACT

PURPOSE: The purposes of this study is to develop an educational program to prevent sexual abuse of children and to improve the physical and mental health of children by providing a rape-free environment and safety education. This program will provide parents and children with information on how to prevent sexual abuse in children. Children learn specific methods to avoid being victimized both at home and outside the home through a learning game and simulation, which is based on problem solving. METHOD: This program was developed based on a literature reviews, surveys and negotiation process. School- aged-children, parents, and teachers were interviewed to reveal their educational needs based on their experiences related to sexual abuse. RESULT: This program includes useful subjects such as safety education, early detection of sexual abuse, crisis management, resource persons, and phone numbers of available hospital. Counseling is provided by researcher or by a pediatric psychiatrist if needed. CONCLUSION: This program could be adequately utilized for prevention of sexual abuse of children. It also will provide an intervention strategy for abused children. This educational program was distributed to all of the elementary school through the Ministry of Education and Human Resources Development.

10.
J Cardiovasc Pharmacol ; 39(2): 271-7, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11791013

ABSTRACT

Daily green tea drinking showed preventive effects on the progression of atherosclerosis. Although epigallocatechin-3-gallate [EGCG] has anti-proliferative effects on various cells, relatively little is known about the molecular mechanisms of the anti-proliferative effects of EGCG. To determine whether the transduction signals and protooncogene expression were affected by EGCG, this study investigated the molecular mechanism of the anti-proliferative effects in basic fibroblast growth factor (bFGF)-stimulated rat aortic smooth muscle cells (RAoSMCs). EGCG inhibited the proliferative response stimulated by 10% fetal bovine serum dose dependently in RAoSMCs (median inhibitory concentration [IC50]: 28.4 x 10(-6) M ). EGCG also inhibited the migration of bFGF-stimulated RAoSMCs in a dose-dependent manner, showing that 21.8 x 10(-6) M of EGCG significantly inhibited the migration by 75 +/- 5% in comparison with bFGF-stimulated migration. In RAoSMCs, EGCG dramatically inhibited Ras activation and c-jun N-terminal kinase (JNK) activity without affecting protein kinase C expression. Induction of c-jun mRNA stimulated by bFGF was significantly reduced dose dependently up to 87.3 x 10(-6) M of EGCG. These results indicate that the anti-proliferative effect of EGCG on RAoSMCs is partly Ras/JNK mediated, independent of protein kinase C, and is attributable to the downregulation of c-jun expression.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Catechin/pharmacology , Fibroblast Growth Factor 2/physiology , Muscle, Smooth/metabolism , Signal Transduction/drug effects , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/metabolism , Blotting, Western , Catechin/analogs & derivatives , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Down-Regulation , Fibroblast Growth Factor 2/pharmacology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth/cytology , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , ras Proteins/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...