Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Inflamm Res ; 70(1): 89-98, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33185705

ABSTRACT

OBJECTIVE: The dysfunction of pulmonary microvascular endothelial cells (PMVECs) is one of the critical characteristics of acute lung injury/acute respiratory distress syndrome (ALI/ARDS) induced by severe infection. PIM1 is a constitutively active serine/threonine kinase that is involved in multiple biological processes. However, the underlying correlation between PIM1 and PMVECs injury remains unclear. The main purpose of this study was to reveal roles of PIM1 and explore the potential mechanisms during the development of endotoxin-induced ALI induced by intraperitoneal LPS administration. MATERIALS AND METHODS: PIM1 level in the lung tissues of endotoxin-induced ALI mice or plasma derived from cardiopulmonary bypass (CPB)-induced ALI patients were measured. The protective roles of PIM1 specific inhibitor SMI-4a on endotoxin-induced lung injuries were evaluated through histological, permeability, neutrophil infiltration and survival assessment. The relationship between PIM1 and ELK3/ICAM-1 axis was validated in vivo and vitro. The correlation between plasma PIM1 and indicative vascular endothelium injury biomarkers (PaO2/FiO2 ratio, Ang-II, E-selectin and PAI-1) levels derived from CPB-induced ALI patient were analyzed. RESULTS: PIM1 expression in the lung tissues was increased in the mice of endotoxin-induced ALI. The PIM1 specific inhibitor SMI-4a administration relieved the severity of endotoxin-induced ALI. More importantly, PIM1 modulates ICAM1 expression through regulating transcription factor ELK3 expression in vitro. Eventually, plasma PIM1 level was positively correlated with Ang-II and PAI-1 levels but negatively correlated with SpO2/FiO2 ratio among CPB induced ALI patients. CONCLUSION: Our results indicated that PIM1 inhibition carried a protective role against endotoxin-induced ALI by modulating the ELK3/ICAM1 axis on PMVECs. PIM1 may be a potential therapeutic target for endotoxin-induced ALI.


Subject(s)
Acute Lung Injury/immunology , Endothelial Cells/immunology , Intercellular Adhesion Molecule-1/immunology , Lung/immunology , Proto-Oncogene Proteins c-ets/immunology , Proto-Oncogene Proteins c-pim-1/immunology , Acute Lung Injury/chemically induced , Animals , Cardiopulmonary Bypass/adverse effects , Cells, Cultured , Humans , Lipopolysaccharides , Lung/cytology , Male , Mice, Inbred C57BL , Microvessels/cytology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/blood
3.
Nat Commun ; 10(1): 1844, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015445

ABSTRACT

Lentiviruses have evolved to acquire an auxiliary protein Vpx to counteract the intrinsic host restriction factor SAMHD1. Although Vpx is phosphorylated, it remains unclear whether such phosphorylation indeed regulates its activity toward SAMHD1. Here we identify the PIM family of serine/threonine protein kinases as the factors responsible for the phosphorylation of Vpx and the promotion of Vpx-mediated SAMHD1 counteraction. Integrated proteomics and subsequent functional analysis reveal that PIM family kinases, PIM1 and PIM3, phosphorylate HIV-2 Vpx at Ser13 and stabilize the interaction of Vpx with SAMHD1 thereby promoting ubiquitin-mediated proteolysis of SAMHD1. Inhibition of the PIM kinases promotes the antiviral activity of SAMHD1, ultimately reducing viral replication. Our results highlight a new mode of virus-host cell interaction in which host PIM kinases facilitate promotion of viral infectivity by counteracting the host antiviral system, and suggest a novel therapeutic strategy involving restoration of SAMHD1-mediated antiviral response.


Subject(s)
HIV Infections/immunology , HIV-2/immunology , Host-Pathogen Interactions/immunology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Proto-Oncogene Proteins/metabolism , SAM Domain and HD Domain-Containing Protein 1/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Biphenyl Compounds/pharmacology , Cell Line, Tumor , HEK293 Cells , HIV Infections/drug therapy , HIV Infections/virology , Humans , Imidazoles/pharmacology , Immune Tolerance , Molecular Dynamics Simulation , Monocytes , Phosphorylation/immunology , Protein Binding/drug effects , Protein Binding/immunology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/immunology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/immunology , Proteolysis/drug effects , Proteomics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/immunology , Pyridazines/pharmacology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , SAM Domain and HD Domain-Containing Protein 1/chemistry , SAM Domain and HD Domain-Containing Protein 1/immunology , Serine/metabolism , Thiazolidines/pharmacology , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/isolation & purification , Virus Replication/drug effects , Virus Replication/immunology
4.
Arthritis Rheumatol ; 71(8): 1308-1318, 2019 08.
Article in English | MEDLINE | ID: mdl-30791224

ABSTRACT

OBJECTIVE: Lupus nephritis (LN) is a major determinant of morbidity and mortality in systemic lupus erythematosus (SLE). Pim-1 regulates lymphocyte proliferation and activation. The role of Pim-1 in autoimmune disease remains unclear. This study was undertaken to test the hypothesis that inhibition of Pim-1 would have therapeutic potential in patients with LN. METHODS: Pim-1 expression was analyzed in lupus-prone (NZB × NZW)F1 mice (n = 6), human peripheral blood mononuclear cells (PBMCs) from SLE patients (n = 10), and glomeruli from patients with LN (n = 8). The therapeutic effect of the Pim-1 inhibitor AZD1208 was assessed in the same murine lupus model (n = 10 mice per group). In vitro analysis was conducted to explore the mechanisms of action of Pim-1 in mouse and human podocytes after Pim-1 expression had been induced by anti-double-stranded DNA (anti-dsDNA) antibody-positive serum. Finally, MRL/lpr mice were used to confirm the therapeutic effects of Pim-1 inhibition in vivo (n = 10 mice per group). RESULTS: Up-regulation of Pim-1 was seen in renal lysates from diseased (NZB × NZW)F1 mice and in PBMCs from patients with SLE and renal biopsy tissue from patients with LN, relative to their control counterparts (each P < 0.05). The Pim-1 inhibitor AZD1208 reduced the severity of proteinuria, glomerulonephritis, renal immune complex deposits, and serum anti-dsDNA antibody levels, concomitant with the suppression of NFATc1 expression and NLRP3 inflammasome activation, in diseased (NZB × NZW)F1 mice (each P < 0.05 versus controls). Moreover, in mouse and human podocytes, Pim-1 knockdown with targeted small interfering RNA (siRNA) suppressed NFATc1 and NLRP3 inflammasome signaling in the presence of anti-dsDNA-positive serum (each P < 0.05 versus control siRNA). Mechanistically, Pim-1 modulated NLRP3 inflammasome activation through intracellular Ca2+ (P < 0.05 versus normal controls). The therapeutic effect of Pim-1 blockade was replicated in MRL/lpr mice. CONCLUSION: These data identify Pim-1 as a critical regulator of LN pathogenesis in patients with SLE. Targeting of the Pim-1/NFATc1/NLRP3 pathway might therefore have therapeutic potential in human LN.


Subject(s)
Cell Proliferation/drug effects , Lupus Erythematosus, Systemic/complications , Lupus Nephritis/drug therapy , Lymphocyte Activation/drug effects , Proto-Oncogene Proteins c-pim-1/pharmacology , Animals , Antibodies, Antinuclear/immunology , Disease Models, Animal , Humans , Inflammasomes/drug effects , Kidney/cytology , Kidney Glomerulus/metabolism , Leukocytes, Mononuclear , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Mice , Mice, Inbred MRL lpr , NFATC Transcription Factors/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Podocytes/metabolism , Proto-Oncogene Proteins c-pim-1/immunology , Signal Transduction/drug effects
5.
Fish Shellfish Immunol ; 74: 491-500, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29355758

ABSTRACT

The Pim1 serine/threonine kinase is associated with multiple cellular functions including proliferation, survival, differentiation, apoptosis, tumorigenesis, immune regulation and inflammation in vertebrates. However, little is known about the role of Pim1 in invertebrate immunity. In this study, we identified and characterized for the first time, a Pim1 (LvPim1) gene in Litopenaeus vannamei, with a full-length cDNA of 2352 bp and a 1119 bp open reading frame (ORF) encoding a putative protein of 372 amino acids, which contains a typical serine/threonine kinase domain. Sequence and phylogenetic analysis revealed that LvPim1 shared a close evolutionary relationship with Pim1 from vertebrates. Real-time qPCR analysis showed that LvPim1 was widely expressed in all tissues tested; with its transcript level induced in hepatopancreas and hemocytes upon challenge with Vibrio parahaemolyticus, Streptoccocus iniae, lipopolysaccharide (LPS), and white spot syndrome virus (WSSV), thus, suggesting its probable involvement in shrimp immune response. Moreover, knockdown of LvPim1 resulted in increased hemocytes apoptosis; shown by high caspase3/7 activity, coupled with increase in pro-apoptotic LvCaspase3 and LvCytochrome C, and decrease in pro-survival LvBcl2, LvIAP1, and LvIAP2 mRNA expression in hemocytes. Finally, LvPim1 knockdown renders shrimps more susceptible to V. parahaemolyticus infection. Taken together, our present data strongly suggest that LvPim1 is involved in modulating shrimp resistance to pathogen infection, promote hemocytes survival, and therefore plays a role in shrimp immune response.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate/genetics , Penaeidae/genetics , Penaeidae/immunology , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/immunology , Amino Acid Sequence , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Base Sequence , Gene Expression Profiling , Lipopolysaccharides/pharmacology , Phylogeny , Proto-Oncogene Proteins c-pim-1/chemistry , Sequence Alignment , Streptococcus iniae/physiology , Vibrio parahaemolyticus/physiology , White spot syndrome virus 1/physiology
6.
Proteomics ; 16(8): 1280-90, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26915739

ABSTRACT

Humoral response in cancer patients can be used for early cancer detection. By screening high-density protein microarrays with sera from colorectal cancer (CRC) patients and controls, we identified 16 tumor-associated antigens (TAAs) exhibiting high diagnostic value. This high number of TAAs requires the development of multiplex assays combining different antigens for a faster and more accurate prediction of CRC. Here, we have developed and optimized a bead-based assay using nine selected TAAs and two controls to provide a multiplex test for early CRC diagnosis. We screened a collection of 307 CRC patients' and control sera with the beads assay to identify and validate the best TAA combination for CRC detection. The multiplex bead-based assay exhibited a similar diagnostic performance to detect the humoral response in comparison to multiple ELISA analyses. After multivariate analysis, a panel composed of GTF2B, EDIL3, HCK, PIM1, STK4, and p53, together with gender and age, was identified as the best combination of TAAs for CRC diagnosis, achieving an AUC of 89.7%, with 66% sensitivity at 90.0% fixed specificity. The model was validated using bootstrapping analysis. In summary, we have developed a novel multiplex bead assay that after validation with a larger independent cohort of sera could be utilized in a high-throughput manner for population screening to facilitate the detection of early CRC patients.


Subject(s)
Antigens, Neoplasm/metabolism , Autoantibodies/metabolism , Colorectal Neoplasms/metabolism , Microspheres , Protein Array Analysis/methods , Aged , Antigens, Neoplasm/immunology , Autoantibodies/blood , Autoantibodies/immunology , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Calcium-Binding Proteins , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell Adhesion Molecules , Colorectal Neoplasms/blood , Colorectal Neoplasms/diagnosis , Enzyme-Linked Immunosorbent Assay , Female , Humans , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Multivariate Analysis , Prognosis , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-hck/immunology , Proto-Oncogene Proteins c-hck/metabolism , Proto-Oncogene Proteins c-pim-1/immunology , Proto-Oncogene Proteins c-pim-1/metabolism , Reproducibility of Results , Sensitivity and Specificity , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
7.
Int Immunopharmacol ; 28(2): 859-65, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25870037

ABSTRACT

Kaempferol is a natural flavonoid found in many vegetables and fruits. Epidemiologic studies have described that Kaempferol intake could reduce risk of cancer, especially lung, gastric, pancreatic and ovarian cancers. Recent studies have shown that Kaempferol could also be beneficial to the body to defend against inflammation, and infection by bacteria and viruses; however, the molecular mechanism of its immunoregulatory function remains largely unknown. Through screening a small molecule library of traditional Chinese medicine (TCM), we identified that Kaempferol could enhance the suppressive function of regulatory T cells (Tregs). Kaempferol was found to increase FOXP3 expression level in Treg cells and prevent pathological symptoms of collagen-induced arthritis in a rat animal model. Kaempferol could also reduce PIM1-mediated FOXP3 phosphorylation at S422. Our study reveals a molecular mechanism that underlies the anti-inflammatory action of Kaempferol for the prevention and treatment of inflammatory diseases such as rheumatoid arthritis, systemic lupus erythematosus, and ankylosing spondylitis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Rheumatoid/immunology , Forkhead Transcription Factors/immunology , Kaempferols/pharmacology , T-Lymphocytes, Regulatory/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Female , Forkhead Transcription Factors/genetics , HEK293 Cells , Humans , Jurkat Cells , Kaempferols/therapeutic use , Phosphorylation/drug effects , Proto-Oncogene Proteins c-pim-1/immunology , RNA, Messenger/metabolism , Rats, Wistar , T-Lymphocytes, Regulatory/immunology
8.
J Biol Chem ; 289(39): 26872-26881, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25096571

ABSTRACT

Previous reports have suggested that human CD4(+) CD25(hi)FOXP3(+) T regulatory cells (Tregs) have functional plasticity and may differentiate into effector T cells under inflammation. The molecular mechanisms underlying these findings remain unclear. Here we identified the residue serine 422 of human FOXP3 as a phosphorylation site that regulates its function, which is not present in murine Foxp3. PIM1 kinase, which is highly expressed in human Tregs, was found to be able to interact with and to phosphorylate human FOXP3 at serine 422. T cell receptor (TCR) signaling inhibits PIM1 induction, whereas IL-6 promotes PIM1 expression in in vitro expanded human Tregs. PIM1 negatively regulates FOXP3 chromatin binding activity by specifically phosphorylating FOXP3 at Ser(422). Our data also suggest that phosphorylation of FOXP3 at the Ser(418) site could prevent FOXP3 phosphorylation at Ser(422) mediated by PIM1. Knockdown of PIM1 in in vitro expanded human Tregs promoted FOXP3-induced target gene expression, including CD25, CTLA4, and glucocorticoid-induced tumor necrosis factor receptor (GITR), or weakened FOXP3-suppressed IL-2 gene expression and enhanced the immunosuppressive activity of Tregs. Furthermore, PIM1-specific inhibitor boosted FOXP3 DNA binding activity in in vitro expanded primary Tregs and also enhanced their suppressive activity toward the proliferation of T effector cells. Taken together, our findings suggest that PIM1 could be a new potential therapeutic target in the prevention and treatment of human-specific autoimmune diseases because of its ability to modulate the immunosuppressive activity of human Tregs.


Subject(s)
Forkhead Transcription Factors/immunology , Proto-Oncogene Proteins c-pim-1/immunology , T-Lymphocytes, Regulatory/immunology , CTLA-4 Antigen/biosynthesis , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Cell Proliferation/physiology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Glucocorticoid-Induced TNFR-Related Protein/biosynthesis , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/immunology , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interleukin-2/biosynthesis , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Jurkat Cells , Phosphorylation/physiology , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Serine/genetics , Serine/immunology , Serine/metabolism , Signal Transduction/physiology , T-Lymphocytes, Regulatory/metabolism
9.
Eur J Immunol ; 42(10): 2544-50, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23042651

ABSTRACT

Basophils have recently been recognized as critical effector cells in allergic reactions and protective immunity against helminths. Precise characterization of basophil biology could help to develop specific therapies that interfere with differentiation, tissue recruitment, or induction of effector functions and thereby ameliorate allergic disorders. The development, homeostasis, and effector functions of basophils are tightly regulated by extrinsic signals and in particular by cytokines. IL-3, GM-CSF, and thymic stromal lymphopoietin activate the STAT5 pathway that promotes proliferation, activation, and cytokine secretion but also induces a negative feedback loop via Pim-1 and SOCS proteins. Basophils further express receptors for IL-18 and IL-33, which are associated with the signaling adaptor MyD88 and activate the NF-κB and MAP kinase pathways. This review focuses on positive and negative regulation of basophils by these cytokines.


Subject(s)
Basophils/immunology , Cytokines/immunology , Immunomodulation , Animals , Cell Differentiation , Cell Proliferation , Humans , NF-kappa B/immunology , Proto-Oncogene Proteins c-pim-1/immunology , STAT5 Transcription Factor/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/immunology
10.
Eur J Immunol ; 42(2): 522-32, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22101984

ABSTRACT

The proto-oncogenes Myc and Pim1, which are deregulated in many types of cancers, are known to cooperate in B lymphoma development. Here we show that overexpression of retrovirally transduced, doxycycline-inducible Myc alone in IL-7-deprived, growth-arrested pre-B cells enhanced cell cycle entry without impairing apoptosis. Overexpression of Pim1 decreased apoptosis, but had no effect on cell cycle entry. Co-expression of Pim1 and Myc inhibited apoptosis and led to IL-7-independent proliferation of the transduced pre-B cells in vitro, while blocking their differentiation to IgM(+) immature cells. Transplantation of Pim1/Myc overexpressing pre-BI cells into B-cell-deficient mice expanded the pre-B-cell compartments up to 100-fold within 4-8 weeks. Transformation remained dependent on the expression of both oncogenes, as removal of doxycycline in vitro and in vivo terminated proliferation and induced differentiation to IgM(+) B cells. In contrast, Pim1/Myc-transduced mature B cells that developed from the oncogene-transduced pre-BI cells in the absence of oncogene overexpression in vivo were not capable of long-term proliferation after induction of Pim and Myc overexpression, neither in vivo nor in vitro, neither with nor without stimulation by polyclonal activators.


Subject(s)
B-Lymphocytes/metabolism , Precursor Cells, B-Lymphoid/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Retroviridae/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , B-Lymphocytes/transplantation , Cell Cycle/genetics , Cell Cycle/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Growth Processes/genetics , Cell Growth Processes/immunology , Cell Line , Cell Transformation, Viral , Cell Transplantation , Doxycycline/administration & dosage , Genetic Vectors/genetics , Homeodomain Proteins/genetics , Interleukin-7/immunology , Interleukin-7/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/pathology , Precursor Cells, B-Lymphoid/transplantation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/immunology , Transgenes/genetics
11.
Cell Immunol ; 272(2): 200-13, 2012.
Article in English | MEDLINE | ID: mdl-22078270

ABSTRACT

PIM kinases are a family of three serine/threonine kinases expressed following T cell activation. Using potent selective small molecule antagonists of PIM-1/3 kinases, we demonstrate a potential role for these enzymes in naïve and effector CD4+ T cell activation. PIM-1/3 inhibition prevented CD4+ T cell proliferation by inducing a G0/G1 cell cycle arrest without affecting cellular survival. In the absence of PIM-1/3 kinase activity, naïve CD4+ T cells failed to fully differentiate into effector cells both in vitro and in vivo. Therapeutic dosing of a PIM-1/3 inhibitor was efficacious in a CD4+ T cell-mediated model of inflammatory bowel disease suggesting that PIM-1 and PIM-3 kinase activity contributes to sustained disease severity. These results demonstrate that PIM-1/3 kinases have an important role in CD4+ T cell responses and inhibition of this activity may provide a therapeutic benefit in T cell-mediated diseases.


Subject(s)
CD4-Positive T-Lymphocytes/enzymology , Inflammatory Bowel Diseases/enzymology , Proto-Oncogene Proteins c-pim-1/immunology , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Growth Processes/immunology , Cell Survival/drug effects , Cytokines/biosynthesis , G1 Phase/drug effects , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Janus Kinases/metabolism , Lymphocyte Activation , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/genetics , Resting Phase, Cell Cycle/drug effects , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Cell Immunol ; 272(1): 87-93, 2011.
Article in English | MEDLINE | ID: mdl-21974958

ABSTRACT

Pim-1 is an important signaling molecule mediating cell proliferation and survival. Our previous study identified a Pim-1 specific monoclonal antibody, P9, with significant inhibitory effect on cell proliferation. Herein, we report that P9 inhibited the activation and proliferation of PHA-stimulated human PBMC and induced them to undergo apoptosis. In contrast, P9 showed little effect on freshly isolated human blood T lymphocytes which poorly expressed Pim-1. P9 also detected an up-regulation of Pim-1 in mouse lymphocytes after mitogen stimulation, and showed similar selective inhibition on stimulated cells as observed with hPBMC. Furthermore, P9 inhibited the in vitro mixed lymphocyte reaction and P9 treatment significantly prolonged the survival of mouse skin allografts (P<0.001). It is concluded that Pim-1 expression correlates with lymphocyte proliferation and activation. P9 functions as a Pim-1 antagonist and is potential for immunosuppressive therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Graft Survival/drug effects , Immunosuppressive Agents/pharmacology , Proto-Oncogene Proteins c-pim-1 , Skin Transplantation/methods , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Apoptosis/drug effects , Cell Proliferation/drug effects , Flow Cytometry , Humans , Immunosuppressive Agents/immunology , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Mice , Phytohemagglutinins/immunology , Phytohemagglutinins/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/immunology , Proto-Oncogene Proteins c-pim-1/metabolism , Skin Transplantation/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transplantation, Homologous
13.
J Immunol ; 185(12): 7460-6, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21068407

ABSTRACT

Pim kinases are emerging as important mediators of cytokine signaling pathways in hematopoietic cells. In this study, we demonstrate that Pim-1 positively regulates RANKL-induced osteoclastogenesis and that Pim-1 expression can be upregulated by RANKL signaling during osteoclast differentiation. The silencing of Pim-1 by RNA interference or overexpression of a dominant negative form of Pim-1 (Pim-1 DN) in bone marrow-derived macrophage cells attenuates RANKL-induced osteoclast formation. Overexpression of Pim-1 DN blocks RANKL-induced activation of TGF-ß-activated kinase 1 (TAK1) and NF-κB as well as expression of NFATc1 during osteoclastogenesis. However, we found that overexpression of TAK1 in the presence of Pim-1 DN rescues NF-κB activation. Additionally, Pim-1 interacts with RANK as well as TAK1, indicating that Pim-1 is involved in RANKL-induced NF-κB activation via TAK1. Furthermore, we demonstrate that Pim-1 also regulates NFATc1 transcription activity and subsequently induces osteoclast-associated receptor expression, an osteoclast-specific gene. Taken together, our results reveal that Pim-1 positively regulates RANKL-induced osteoclastogenesis.


Subject(s)
Cell Differentiation/immunology , NF-kappa B/immunology , NFATC Transcription Factors/immunology , Osteoclasts/immunology , Proto-Oncogene Proteins c-pim-1/immunology , RANK Ligand/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cells, Cultured , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , MAP Kinase Kinase Kinases/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred ICR , NF-kappa B/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , RNA Interference/immunology , Transcription, Genetic/immunology , Up-Regulation/genetics , Up-Regulation/immunology
14.
Exp Hematol ; 37(11): 1284-94, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19703513

ABSTRACT

OBJECTIVE: Based on our previous findings that Pim-1 was expressed on the cell surface and could be targeted with a highly specific anti-Pim-1 monoclonal antibody (P9), this study aims to evaluate the possibility that Pim-1 could be targeted for the treatment of human leukemia. MATERIALS AND METHODS: Pim-1 expression was investigated in a series of human leukemia cell lines with immunohistochemistry and flow cytometry. The inhibitory effect of P9 on cell proliferation was evaluated with (3)H-thymidine incorporation assay. Cell apoptosis was assayed with Annexin-V/propidium iodide dual staining. The in vivo effect of P9 was evaluated with xenograft tumor models in severe combined immunodeficient mice. RESULTS: Pim-1 expression varied depending on the cell lines and correlated with the inhibitory effects mediated by P9. An association between Pim-1 expression and drug resistance was observed. Although the drug-resistant CEM/A7R cells were highly resistant to cytotoxic P-glycoprotein substrates, their growth was inhibited by P9 as demonstrated by in vitro proliferation assay and in vivo inhibition of xenograft tumors. P9 had little effect on P-glycoprotein expression and intracellular Rhodamine 123 accumulation, but it inhibited the phosphorylation of Bad and induced apoptosis. CONCLUSIONS: Pim-1 is variably expressed in leukemia cell lines and associated with drug resistance. Targeting Pim-1 with monoclonal antibody could be explored for the treatment of leukemia and may represent a novel strategy to overcome drug resistance.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Leukemia/pathology , Neoplasm Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/therapeutic use , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia, Experimental/drug therapy , Mice , Mice, SCID , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-pim-1/biosynthesis , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/immunology , Tumor Stem Cell Assay , Verapamil/pharmacology , Xenograft Model Antitumor Assays , bcl-Associated Death Protein/metabolism
15.
J Allergy Clin Immunol ; 123(3): 603-11, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19152965

ABSTRACT

BACKGROUND: Eosinophil differentiation, activation, and survival are largely regulated by IL-5. IL-5-mediated transmembrane signal transduction involves both Lyn-mitogen-activated protein kinases and Janus kinase 2-signal transducer and activator of transcription pathways. OBJECTIVE: We sought to determine whether additional signaling molecules/pathways are critically involved in IL-5-mediated eosinophil survival. METHODS: Eosinophil survival and apoptosis were measured in the presence and absence of IL-5 and defined pharmacologic inhibitors in vitro. The specific role of the serine/threonine kinase proviral integration site for Moloney murine leukemia virus (Pim) 1 was tested by using HIV-transactivator of transcription fusion proteins containing wild-type Pim-1 or a dominant-negative form of Pim-1. The expression of Pim-1 in eosinophils was analyzed by means of immunoblotting and immunofluorescence. RESULTS: Although pharmacologic inhibition of phosphatidylinositol-3 kinase (PI3K) by LY294002, wortmannin, or the selective PI3K p110delta isoform inhibitor IC87114 was successful in each case, only LY294002 blocked increased IL-5-mediated eosinophil survival. This suggested that LY294002 inhibited another kinase that is critically involved in this process in addition to PI3K. Indeed, Pim-1 was rapidly and strongly expressed in eosinophils after IL-5 stimulation in vitro and readily detected in eosinophils under inflammatory conditions in vivo. Moreover, by using specific protein transfer, we identified Pim-1 as a critical element in IL-5-mediated antiapoptotic signaling in eosinophils. CONCLUSIONS: Pim-1, but not PI3K, plays a major role in IL-5-mediated antiapoptotic signaling in eosinophils.


Subject(s)
Apoptosis , Eosinophils/immunology , Interleukin-5/immunology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-pim-1/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Androstadienes/pharmacology , Cells, Cultured , Chromones/pharmacology , Eosinophils/drug effects , Eosinophils/enzymology , Humans , Hypersensitivity/enzymology , Hypersensitivity/immunology , Interleukin-5/pharmacology , Janus Kinase 2/immunology , Janus Kinase 2/metabolism , Microscopy, Confocal , Morpholines/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein , Phosphatidylinositol 3-Kinases/immunology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/immunology , Quinazolines/pharmacology , Tyrphostins/pharmacology , Wortmannin , Xanthenes/pharmacology
16.
J Immunol Methods ; 316(1-2): 8-17, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16989855

ABSTRACT

The presence of neutralizing antibodies against protein therapeutics is a concern in the biomedical field. Such antibodies not only reduce the efficacy of protein therapeutics, but also impose potential dangers to the patients receiving them. To date, a small number of in vitro cell-based bioassays for detecting neutralizing antibodies against therapeutic proteins have been developed. Most of the existing assays, however, either involve the use of radioactive materials or have limited sensitivities and/or poor specificities. With advances in mRNA profiling and detection techniques, we have established a novel and non-radioactive bioassay system using branched DNA (bDNA) technology for detecting protein-therapeutic neutralizing antibodies in patient serum. Our assay measures the variations of target gene expression that reflect the biologic effect of the therapeutic agent and the capability of the antibodies, if present, to neutralize the therapeutics. Compared with most existing assays, the new assay is more sensitive and specific, and completely eliminates the use of radioactive materials. Application of the new assay system can be widely expanded if new target genes and responding cell lines for other therapeutics are identified or engineered.


Subject(s)
Antibodies/blood , Branched DNA Signal Amplification Assay/methods , Oligonucleotide Array Sequence Analysis/methods , Proto-Oncogene Proteins c-pim-1/immunology , Antibodies/genetics , Antibody Specificity , Butadienes/pharmacology , Cell Line, Tumor , Chromones/pharmacology , Erythropoietin/immunology , Erythropoietin/pharmacology , Humans , Morpholines/pharmacology , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/biosynthesis , Proto-Oncogene Proteins c-pim-1/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...