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










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 315(3): H563-H570, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29949382

ABSTRACT

Cardiac fibroblasts are critical mediators of fibrotic remodeling in the failing heart and transform into myofibroblasts in the presence of profibrotic factors such as transforming growth factor-ß. Myocardial fibrosis worsens cardiac function, accelerating the progression to decompensated heart failure (HF). We investigated the effects of a novel inhibitor (NM922; NovoMedix, San Diego, CA) of the conversion of normal fibroblasts to the myofibroblast phenotype in the setting of pressure overload-induced HF. NM922 inhibited fibroblast-to-myofibroblast transformation in vitro via a reduction of activation of the focal adhesion kinase-Akt-p70S6 kinase and STAT3/4E-binding protein 1 pathways as well as via induction of cyclooxygenase-2. NM922 preserved left ventricular ejection fraction ( P < 0.05 vs. vehicle) and significantly attenuated transverse aortic constriction-induced LV dilation and hypertrophy ( P < 0.05 compared with vehicle). NM922 significantly ( P < 0.05) inhibited fibroblast activation, as evidenced by reduced myofibroblast counts per square millimeter of tissue area. Picrosirius red staining demonstrated that NM922 reduced ( P < 0.05) interstitial fibrosis compared with mice that received vehicle. Similarly, NM922 hearts had lower mRNA levels ( P < 0.05) of collagen types I and III, lysyl oxidase, and TNF-α at 16 wk after transverse aortic constriction. Treatment with NM922 after the onset of cardiac hypertrophy and HF resulted in attenuated myocardial collagen formation and adverse remodeling with preservation of left ventricular ejection fraction. Future studies are aimed at further elucidation of the molecular and cellular mechanisms by which this novel antifibrotic agent protects the failing heart. NEW & NOTEWORTHY Our data demonstrated that a novel antifibrotic agent, NM922, blocks the activation of fibroblasts, reduces the formation of cardiac fibrosis, and preserves cardiac function in a murine model of heart failure with reduced ejection fraction.


Subject(s)
Cardiotonic Agents/pharmacology , Heart Failure/drug therapy , Myofibroblasts/drug effects , Sulfonamides/pharmacology , Ventricular Remodeling/drug effects , Animals , Cardiotonic Agents/therapeutic use , Cells, Cultured , Collagen/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism , Protein-Lysine 6-Oxidase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , STAT3 Transcription Factor/metabolism , Sulfonamides/chemistry , Sulfonamides/therapeutic use , Transforming Growth Factor beta/metabolism
2.
Nat Struct Mol Biol ; 21(9): 803-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108355

ABSTRACT

The Cul4-Rbx1-DDB1-Cereblon E3 ubiquitin ligase complex is the target of thalidomide, lenalidomide and pomalidomide, therapeutically important drugs for multiple myeloma and other B-cell malignancies. These drugs directly bind Cereblon (CRBN) and promote the recruitment of substrates Ikaros (IKZF1) and Aiolos (IKZF3) to the E3 complex, thus leading to substrate ubiquitination and degradation. Here we present the crystal structure of human CRBN bound to DDB1 and the drug lenalidomide. A hydrophobic pocket in the thalidomide-binding domain (TBD) of CRBN accommodates the glutarimide moiety of lenalidomide, whereas the isoindolinone ring is exposed to solvent. We also solved the structures of the mouse TBD in the apo state and with thalidomide or pomalidomide. Site-directed mutagenesis in lentiviral-expression myeloma models showed that key drug-binding residues are critical for antiproliferative effects.


Subject(s)
Angiogenesis Inhibitors/pharmacology , DNA-Binding Proteins/metabolism , Peptide Hydrolases/metabolism , Thalidomide/analogs & derivatives , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Angiogenesis Inhibitors/chemistry , Animals , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , Humans , Lenalidomide , Mice , Molecular Docking Simulation , Molecular Sequence Data , Peptide Hydrolases/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Thalidomide/chemistry , Thalidomide/pharmacology , Ubiquitin-Protein Ligases
3.
Blood ; 124(10): 1637-44, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-24990888

ABSTRACT

Lenalidomide has demonstrated clinical activity in patients with chronic lymphocytic leukemia (CLL), even though it is not cytotoxic for primary CLL cells in vitro. We examined the direct effect of lenalidomide on CLL-cell proliferation induced by CD154-expressing accessory cells in media containing interleukin-4 and -10. Treatment with lenalidomide significantly inhibited CLL-cell proliferation, an effect that was associated with the p53-independent upregulation of the cyclin-dependent kinase inhibitor, p21(WAF1/Cip1) (p21). Silencing p21 with small interfering RNA impaired the capacity of lenalidomide to inhibit CLL-cell proliferation. Silencing cereblon, a known molecular target of lenalidomide, impaired the capacity of lenalidomide to induce expression of p21, inhibit CD154-induced CLL-cell proliferation, or enhance the degradation of Ikaros family zinc finger proteins 1 and 3. We isolated CLL cells from the blood of patients before and after short-term treatment with low-dose lenalidomide (5 mg per day) and found the leukemia cells were also induced to express p21 in vivo. These results indicate that lenalidomide can directly inhibit proliferation of CLL cells in a cereblon/p21-dependent but p53-independent manner, at concentrations achievable in vivo, potentially contributing to the capacity of this drug to inhibit disease-progression in patients with CLL.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Peptide Hydrolases/physiology , Thalidomide/analogs & derivatives , Adaptor Proteins, Signal Transducing , Gene Expression Regulation, Leukemic/drug effects , HeLa Cells , Humans , Lenalidomide , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Signal Transduction/drug effects , Thalidomide/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/physiology , Ubiquitin-Protein Ligases
4.
Blood ; 118(4): 1109-12, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21536862

ABSTRACT

Pharmacologic induction of fetal hemoglobin (HbF) expression is an effective treatment strategy for sickle cell disease (SCD) and ß-thalassemia. Pomalidomide is a potent structural analog of thalidomide and member of a new class of immunomodulatory drugs. Recent reports demonstrated that pomalidomide reduced or eliminated transfusion requirements in certain hematologic malignancies and induced HbF ex vivo in CD34(+) progenitor cells from healthy and SCD donors. We investigated the effects of pomalidomide on erythropoiesis and hemoglobin synthesis in a transgenic mouse model of SCD. We found that 8 weeks of treatment with pomalidomide induced modest increases of HbF with similar efficacy as hydroxyurea. However, in stark contrast to hydroxyurea's myelosuppressive effects, pomalidomide augmented erythropoiesis and preserved bone marrow function. Surprisingly, combinatory therapy with both drugs failed to mitigate hydroxyurea's myelotoxic effects and caused loss of HbF induction. These findings support further evaluation of pomalidomide as a novel therapy for SCD.


Subject(s)
Anemia, Sickle Cell/blood , Antisickling Agents/pharmacology , Bone Marrow/drug effects , Erythropoiesis/drug effects , Fetal Hemoglobin/drug effects , Thalidomide/analogs & derivatives , Animals , Disease Models, Animal , Hydroxyurea/adverse effects , Mice , Mice, Knockout , Mice, Transgenic , Thalidomide/pharmacology
5.
Blood ; 114(2): 338-45, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19417207

ABSTRACT

IMiDs immunomodulatory drugs, including lenalidomide and pomalidomide represent a novel class of small molecule anticancer and anti-inflammatory drugs with broad biologic activities. However, the molecular mechanism through which these drugs exert their effects is largely undefined. Using pomalidomide and primary human monocytes, we report that pomalidomide rapidly and selectively activated RhoA and Rac1, but not Cdc42 or Ras, in the absence of any costimulation. Consistent with the activation of Rho GTPases, we found that pomalidomide enhanced F-actin formation, stabilized microtubules, and increased cell migration, all of which were blocked by selective inhibitors of ROCK1 and Rac1. Further, we showed that in Swiss 3T3 cells, pomalidomide only activated RhoA, not Rac1 or Cdc42, and potently induced stress fiber formation. The pomalidomide effect on actin cytoskeleton was blocked by the ROCK1 inhibitor, but not Rac1 inhibitor. Finally, we demonstrated that pomalidomide was able to regulate the activity of Rho GTPases and the formation of F-actin in primary human T cells as it did in monocytes and showed that the activation of RhoA was essential for pomalidomide-induced interleukin-2 expression in T cells. These novel activities provide what we believe a critical mechanism by which IMiDs drugs function as therapeutic immunomodulatory agents.


Subject(s)
Cytoskeleton/drug effects , Cytoskeleton/enzymology , Immunosuppressive Agents/pharmacology , Thalidomide/analogs & derivatives , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , Cytoskeleton/immunology , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/metabolism , Mice , Monocytes/cytology , Monocytes/drug effects , Monocytes/enzymology , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Thalidomide/pharmacology
6.
J Med Chem ; 52(6): 1522-4, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19256507

ABSTRACT

In this communication, we report the discovery of 1S (apremilast), a novel potent and orally active phosphodiesterase 4 (PDE4) and tumor necrosis factor-alpha inhibitor. The optimization of previously reported 3-(1,3-dioxo-1,3-dihydroisoindol-2-yl)-3-(3,4-dimethoxyphenyl)propionic acid PDE4 inhibitors led to this series of sulfone analogues. Evaluation of the structure-activity relationship of substitutions on the phthalimide group led to the discovery of an acetylamino analogue 1S, which is currently in clinical trials.


Subject(s)
Drug Discovery , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Thalidomide/analogs & derivatives , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Administration, Oral , Animals , Humans , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/chemistry , Rats , Stereoisomerism , Structure-Activity Relationship , Thalidomide/administration & dosage , Thalidomide/chemistry , Thalidomide/pharmacology
7.
Cancer Immunol Immunother ; 57(12): 1849-59, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18392823

ABSTRACT

Revlimid (Lenalidomide, CC-5013) and CC-4047 are IMiDs immunomodulatory drugs that have been described as having immunomodulatory properties and anti-tumor activity. Here we report proapoptotic effects of CC-5013 and CC-4047 on tumor cells in a co-culture model of PBMC and tumor cells. CC-5013 and CC-4047 enhanced PBMC activity leading to tumor cell apoptosis in K562/PBMC co-culture model. We also demonstrate that the natural killer (NK) cell population of PBMC was essential in inducing K562 apoptosis. Increases of NK and natural killer T (NKT) cell populations by CC-5013 and CC-4047 was observed along with modulation of NK cell CD56 adhesion marker. In addition, our data indicate that NK activation by CC-4047 was dependent on other cell types of PBMC. We expanded the application of K562/PBMC co-culture model to other hematological and solid tumors. In Raji/PBMC co-culture model, CC-5013 and CC-4047 dose-dependently augmented tumor cell apoptosis. Pre-treatment of Raji cells with Rituximab further enhanced apoptosis induced by CC-5013 or CC-4047-treated PBMC. Moreover, CC-5013 and CC-4047 significantly increased PC-3 prostate cancer cell apoptosis in PC-3/PBMC co-culture, either as single agent or in combination with Docetaxel. Together, the results reveal that co-culture models are suitable cellular systems to assess anti-tumor activities of these compounds. Our findings support clinical evaluation of CC-5013 and CC-4047 in relapsed NHL with Rituximab and in prostate cancer with Docetaxel.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Killer Cells, Natural/drug effects , Neoplasms/immunology , Thalidomide/analogs & derivatives , Cell Line, Tumor , Coculture Techniques , Drug Screening Assays, Antitumor/methods , Flow Cytometry , Humans , Killer Cells, Natural/immunology , Lenalidomide , Leukocytes, Mononuclear/immunology , Lymphoma, Non-Hodgkin/immunology , Male , Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Thalidomide/pharmacology
8.
Blood ; 111(9): 4690-9, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18305219

ABSTRACT

Decreased p27(Kip1) levels are a poor prognostic factor in many malignancies, and can occur through up-regulation of SCF(Skp2) E3 ligase function, resulting in enhanced p27 ubiquitination and proteasome-mediated degradation. While proteasome inhibitors stabilize p27(Kip1), agents inhibiting SCF(Skp2) may represent more directly targeted drugs with the promise of enhanced efficacy and reduced toxicity. Using high-throughput screening, we identified Compound A (CpdA), which interfered with SCF(Skp2) ligase function in vitro, and induced specific accumulation of p21 and other SCF(Skp2) substrates in cells without activating a heat-shock protein response. CpdA prevented incorporation of Skp2 into the SCF(Skp2) ligase, and induced G(1)/S cell-cycle arrest as well as SCF(Skp2)- and p27-dependent cell killing. This programmed cell death was caspase-independent, and instead occurred through activation of autophagy. In models of multiple myeloma, CpdA overcame resistance to dexamethasone, doxorubicin, and melphalan, as well as to bortezomib, and also acted synergistically with this proteasome inhibitor. Importantly, CpdA was active against patient-derived plasma cells and both myeloid and lymphoblastoid leukemia blasts, and showed preferential activity against neoplastic cells while relatively sparing other marrow components. These findings provide a rational framework for further development of SCF(Skp2) inhibitors as a novel class of antitumor agents.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p27/physiology , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Cell Line, Tumor , Drug Delivery Systems/methods , Drug Resistance, Neoplasm , Humans , Multiple Myeloma/drug therapy
9.
J Clin Invest ; 118(1): 248-58, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18064299

ABSTRACT

Sickle-cell disease (SCD) and beta thalassemia constitute worldwide public health problems. New therapies, including hydroxyurea, have attempted to augment the synthesis of fetal hemoglobin (HbF) and improve current treatment. Lenalidomide and pomalidomide are members of a class of immunomodulators used as anticancer agents. Because clinical trials have demonstrated that lenalidomide reduces or eliminates the need for transfusions in some patients with disrupted blood cell production, we investigated the effects of lenalidomide and pomalidomide on erythropoiesis and hemoglobin synthesis. We used an in vitro erythropoiesis model derived from human CD34+ progenitor cells from normal and SCD donors. We found that both compounds slowed erythroid maturation, increased proliferation of immature erythroid cells, and regulated hemoglobin transcription, resulting in potent induction of HbF without the cytotoxicity associated with other HbF inducers. When combined with hydroxyurea, pomalidomide and, to a lesser extent, lenalidomide were found to have synergistic effects on HbF upregulation. Our results elucidate what we believe to be a new mechanism of action of pomalidomide and lenalidomide and support the hypothesis that pomalidomide, used alone or in combination with hydroxyurea, may improve erythropoiesis and increase the ratio of fetal to adult hemoglobin. These findings support the evaluation of pomalidomide as an innovative new therapy for beta-hemoglobinopathies.


Subject(s)
Anemia, Sickle Cell/metabolism , Antigens, CD34 , Antineoplastic Agents/pharmacology , Erythropoiesis/drug effects , Fetal Hemoglobin/biosynthesis , Thalidomide/analogs & derivatives , beta-Thalassemia/metabolism , Anemia, Sickle Cell/therapy , Antineoplastic Agents/therapeutic use , Antisickling Agents/pharmacology , Antisickling Agents/therapeutic use , Blood Transfusion , Cells, Cultured , Clinical Trials as Topic , Drug Evaluation, Preclinical , Drug Synergism , Erythroid Cells/metabolism , Humans , Hydroxyurea/agonists , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , Lenalidomide , Models, Biological , Thalidomide/agonists , Thalidomide/pharmacology , Thalidomide/therapeutic use , Up-Regulation/drug effects , beta-Thalassemia/therapy
10.
J Clin Immunol ; 27(2): 210-20, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17308870

ABSTRACT

COX2 (prostaglandin G/H synthase, PTGS2) is a well-validated target in the fields of both oncology and inflammation. Despite their significant toxicity profile, non-steroidal anti-inflammatory drugs (NSAIDs) have become standard of care in the treatment of many COX2-mediated inflammatory conditions. In this report, we show that one IMiDs((R)) immunomodulatory drug, CC-4047, can reduce the levels of COX2 and the production of prostaglandins (PG) in human LPS-stimulated monocytes. The inhibition of COX2 by CC-4047 occurs at the level of gene transcription, by reducing the LPS-stimulated transcriptional activity at the COX2 gene. Because it is a transcriptional rather than an enzymatic inhibitor of COX2, CC-4047 inhibition of PG production is not susceptible to competition by exogenous arachadonic acid (AA). The distinct mechanisms of action allow CC-4047 and a COX2-selective NSAID to work additively to block PG secretion from monocytes. CC-4047 does not, however, block COX2 induction in or prostacyclin secretion from IL-1beta stimulated human umbilical vein endothelial cells (HUVEC) cells, nor does it inhibit COX1 in either monocytes or HUVEC cells. CC-4047 also inhibits COX2 and PG production in monocytes derived from patients with sickle cell disease (SCD). Taken together, the data in this manuscript suggest CC-4047 will provide important anti-inflammatory benefit to patients and will improve the safety of NSAIDs in the treatment of SCD or other inflammatory conditions.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/drug effects , Immunologic Factors/pharmacology , Membrane Proteins/drug effects , Thalidomide/analogs & derivatives , Transcription, Genetic/drug effects , Anemia, Sickle Cell/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blotting, Western , Drug Synergism , Gene Expression/drug effects , Humans , Immunoprecipitation , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Monocytes/metabolism , Nitrobenzenes/pharmacology , Prostaglandins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Thalidomide/pharmacology
11.
Cancer Res ; 67(2): 746-55, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17234786

ABSTRACT

Clinical studies involving patients with myelodysplastic syndromes or multiple myeloma have shown the efficacy of lenalidomide by reducing and often eliminating malignant cells while restoring the bone marrow function. To better understand these clinical observations, we investigated and compared the effects of lenalidomide and a structurally related analogue, CC-4047, on the proliferation of two different human hematopoietic cell models: the Namalwa cancer cell line and normal CD34+ progenitor cells. Both compounds had antiproliferative effects on Namalwa cells and pro-proliferative effects on CD34+ cells, whereas p21WAF-1 expression was up-regulated in both cell types. In Namalwa cells, the up-regulation of p21WAF-1 correlated well with the inhibition of cyclin-dependent kinase (CDK) 2, CDK4, and CDK6 activity leading to pRb hypophosphorylation and cell cycle arrest, whereas in CD34+ progenitor cells the increase of p21WAF-1 did not inhibit proliferation. Similarly, antiproliferation results were observed in two B lymphoma cell lines (LP-1 and U266) but interestingly not in normal B cells where a protection of apoptosis was found. Finally, CC-4047 and lenalidomide had synergistic effects with valproic acid [a histone deacetylase (HDAC) inhibitor] by increasing the apoptosis of Namalwa cells and enhancing CD34+ cell expansion. Our results indicate that lenalidomide and CC-4047 have opposite effects in tumor cells versus normal cells and could explain, at least in part, the reduction of malignant cells and the restoration of bone marrow observed in patients undergoing lenalidomide treatment. Moreover, this study provides new insights on the cellular pathways affected by lenalidomide and CC-4047, proposes new potential clinical uses, such as bone marrow regeneration, and suggests that the combination of lenalidomide or CC-4047 with certain HDAC inhibitors may elevate the therapeutic index in the treatment of hematologic malignancies.


Subject(s)
Antigens, CD34/biosynthesis , Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , Hematopoietic Stem Cells/drug effects , Lymphoma, B-Cell/drug therapy , Thalidomide/analogs & derivatives , B-Lymphocytes/pathology , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Lenalidomide , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Phosphorylation/drug effects , Retinoblastoma Protein/metabolism , Thalidomide/pharmacology , Up-Regulation/drug effects
12.
Bioorg Med Chem Lett ; 13(20): 3415-7, 2003 Oct 20.
Article in English | MEDLINE | ID: mdl-14505639

ABSTRACT

Thalidomide, (1), has made a remarkable comeback from its days of a sedative with teratogenic properties due to its ability to selectively inhibit TNF-alpha, a key pro-inflammatory cytokine and its clinical benefit in the treatment of cancer. Thalidomide contains one chiral center and is known to be chirally unstable under in vitro and in vivo conditions. It has been hypothesized that different biological properties are associated with each isomer. Thus, chirally stable analogues of thalidomide, alpha-fluorothalidomide, (3) and alpha-fluoro-4-aminothalidomide (4) were prepared by electrophilic fluorination. Analogue 3 was found to be cytotoxic and did not inhibit TNF-alpha production in LPS stimulated hPBMC below toxic concentrations. On the other hand, 4 was non-cytotoxic at the tested concentrations and was found to be 830-fold more potent than thalidomide as TNF-alpha inhibitor.


Subject(s)
Fluorine/chemistry , Thalidomide/chemistry , Cells, Cultured , Humans , Monocytes/drug effects , Monocytes/metabolism , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis
13.
J Pharmacol Exp Ther ; 305(3): 1222-32, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12649301

ABSTRACT

CC-4047 (Actimid) and CC-5013 (Revimid) belong to a class of thalidomide analogs collectively known as the immunomodulatory drugs (IMiDs), which are currently being assessed in the treatment of patients with multiple myeloma and other cancers. IMiDs potently enhance T cell and natural killer cell responses and inhibit tumor necrosis factor-alpha, interleukin (IL)-1 beta, and IL-12 production from LPS-stimulated peripheral blood mononuclear cells. However, the molecular mechanism of action for these compounds is unknown. Herein, we report on the ability of the IMiDs to up-regulate production of IL-2 from activated human CD4+ and CD8+ peripheral blood T cells, production of IL-2 and IFN-gamma from T helper (Th)1-type cells, and production of IL-5 and IL-10 from Th2-type cells. Elevation of IL-2 production from Jurkat T cells was observed as early as 6 h poststimulation and correlated with an increase in IL-2 promoter activity that was dependent upon the proximal but not the distal AP-1 binding site. The IMiDs enhanced AP-1-driven transcriptional activity 2- to 4-fold after 6 h of T cell stimulation, and their relative potencies for AP-1 activation correlated with their potencies for increased IL-2 production in Jurkat T cells and in CD4+ or CD8+ human peripheral blood T cells. The most potent of these IMiDs, CC-4047, had no effect on nuclear factor of activated T cells transcriptional activity, calcium signaling, or phosphorylation of extracellular signal-regulated kinase 1/2, c-Jun NH2-terminal kinase 1/2, p38 mitogen-activated protein kinase, or c-Jun/Jun D in Jurkat T cells. These data suggest that IMiDs increase T cell cytokine production by potentiating AP-1 transcriptional activity.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cytokines/biosynthesis , Proto-Oncogene Proteins , T-Lymphocytes/drug effects , Thalidomide/pharmacology , Transcription Factor AP-1/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Calcineurin/metabolism , Calcium Signaling , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Interleukin-2/biosynthesis , JNK Mitogen-Activated Protein Kinases , Janus Kinase 2 , Jurkat Cells , Leukocyte Common Antigens/metabolism , Lymphocyte Activation , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , T-Lymphocytes/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism , Thalidomide/analogs & derivatives , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...