Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Cell Rep ; 38(2): 110215, 2022 01 11.
Article En | MEDLINE | ID: mdl-35021079

Macrophages are known to mediate anti-helminth responses, but it remains uncertain which subsets are involved or how macrophages actually kill helminths. Here, we show rapid monocyte recruitment to the lung after infection with the nematode parasite Nippostrongylus brasiliensis. In this inflamed tissue microenvironment, these monocytes differentiate into an alveolar macrophage (AM)-like phenotype, expressing both SiglecF and CD11c, surround invading parasitic larvae, and preferentially kill parasites in vitro. Monocyte-derived AMs (Mo-AMs) express type 2-associated markers and show a distinct remodeling of the chromatin landscape relative to tissue-derived AMs (TD-AMs). In particular, they express high amounts of arginase-1 (Arg1), which we demonstrate mediates helminth killing through L-arginine depletion. These studies indicate that recruited monocytes are selectively programmed in the pulmonary environment to express AM markers and an anti-helminth phenotype.


Lung/immunology , Macrophages, Alveolar/immunology , Strongylida Infections/immunology , Animals , Arginase/metabolism , Cell Differentiation , Cytokines , Female , Lung/parasitology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Nippostrongylus , Strongylida Infections/parasitology
2.
J Nanobiotechnology ; 13: 21, 2015 Mar 12.
Article En | MEDLINE | ID: mdl-25886153

BACKGROUND: Electrospinning is a simple and effective method for fabricating micro- and nanofiber matrices. Electrospun fibre matrices have numerous advantages for use as tissue engineering scaffolds, such as high surface area-to-volume ratio, mass production capability and structural similarity to the natural extracellular matrix (ECM). Therefore, electrospun matrices, which are composed of biocompatible polymers and various biomaterials, have been developed as biomimetic scaffolds for the tissue engineering applications. In particular, graphene oxide (GO) has recently been considered as a novel biomaterial for skeletal muscle regeneration because it can promote the growth and differentiation of myoblasts. Therefore, the aim of the present study was to fabricate the hybrid fibre matrices that stimulate myoblasts differentiation for skeletal muscle regeneration. RESULTS: Hybrid fibre matrices composed of poly(lactic-co-glycolic acid, PLGA) and collagen (Col) impregnated with GO (GO-PLGA-Col) were successfully fabricated using an electrospinning process. Our results indicated that the GO-PLGA-Col hybrid matrices were comprised of randomly-oriented continuous fibres with a three-dimensional non-woven porous structure. Compositional analysis showed that GO was dispersed uniformly throughout the GO-PLGA-Col matrices. In addition, the hydrophilicity of the fabricated matrices was significantly increased by blending with a small amount of Col and GO. The attachment and proliferation of the C2C12 skeletal myoblasts were significantly enhanced on the GO-PLGA-Col hybrid matrices. Furthermore, the GO-PLGA-Col matrices stimulated the myogenic differentiation of C2C12 skeletal myoblasts, which was enhanced further under the culture conditions of the differentiation media. CONCLUSIONS: Taking our findings into consideration, it is suggested that the GO-PLGA-Col hybrid fibre matrices can be exploited as potential biomimetic scaffolds for skeletal tissue engineering and regeneration because these GO-impregnated hybrid matrices have potent effects on the induction of spontaneous myogenesis and exhibit superior bioactivity and biocompatibility.


Biomimetic Materials/chemistry , Collagen/chemistry , Graphite/chemistry , Lactic Acid/chemistry , Myoblasts/cytology , Polyglycolic Acid/chemistry , Animals , Cell Adhesion , Cell Culture Techniques/methods , Cell Differentiation , Cell Proliferation , Extracellular Matrix/chemistry , Mice , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Muscle, Skeletal/cytology , Polylactic Acid-Polyglycolic Acid Copolymer , Spectroscopy, Fourier Transform Infrared , Surface Properties , Tissue Scaffolds , X-Ray Diffraction
3.
J Nanosci Nanotechnol ; 15(10): 7907-12, 2015 Oct.
Article En | MEDLINE | ID: mdl-26726438

Recently, there has been considerable effort to develop suitable scaffolds for tissue engineering applications. Cell adhesion is a prerequisite for cells to survive. In nature, the extracellular matrix (ECM) plays this role. Therefore, an ideal scaffold should be structurally similar to the natural ECM and have biocompatibility and biodegradability. In addition, the scaffold should have biofunctionality, which provides the potent ability to enhance the cellular behaviors, such as adhesion, proliferation and differentiation. This study concentrates on fabricating cell-adhesive matrices composed of RGD peptide-displaying M13 bacteriophage (RGD-M13 phage) and poly(lactic-co-glycolic acid, PLGA) nanofibers. Long rod-shaped M13 bacteriophages are non-toxic and can express many desired proteins on their surface. A genetically engineered M13 phage was constructed to display RGD peptides on its surface. PLGA is a biodegradable polymer with excellent biocompatibility and suitable physicochemical property for adhesive matrices. In this study, RGD-M13 phage/PLGA hybrid nanofiber matrices were fabricated by electrospinning. The physicochemical properties of these matrices were characterized by scanning electron microscopy, atomic force microscopy, Raman spectroscopy, and contact angle measurement. In addition, the cellular behaviors, such as the initial attachment, proliferation and differentiation, were analyzed by a CCK-8 assay and immunofluorescence staining to evaluate the potential application of these matrices to tissue engineering scaffolds. The RGD-M13 phage/PLGA nanofiber matrices could enhance the cellular behaviors and promote the differentiation of C2C12 myoblasts. These results suggest that the RGD-M13 phage/PLGA nanofiber matrices are beneficial to myoblast differentiation and can serve as effective tissue engineering scaffolds.


Antimicrobial Cationic Peptides , Bacteriophage M13/chemistry , Cell Differentiation/drug effects , Myoblasts, Skeletal/metabolism , Nanofibers/chemistry , Oligopeptides , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Cell Adhesion/drug effects , Cell Line , Mice , Myoblasts, Skeletal/cytology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Tissue Scaffolds/chemistry
4.
J Nanosci Nanotechnol ; 15(10): 7966-70, 2015 Oct.
Article En | MEDLINE | ID: mdl-26726448

Osteoprogenitor cells play a significant role in the growth or repair of bones, and have great potential as cell sources for regenerative medicine and bone tissue engineering, but control of their specific differentiation into bone cells remains a challenge. Graphene-based nanomaterials are attractive candidates for biomedical applications as substrates for stem cell (SC) differentiation, scaffolds in tissue engineering, and components of implant devices owing to their biocompatible, transferable and implantable properties. This study examined the enhanced osteogenic differentiation of human mesenchymal stem cells (hMSCs) by reduced graphene oxide (rGO) nanoparticles (NPs), and rGO NPs was prepared by reducing graphene oxide (GO) with a hydrazine treatment followed by annealing in argon and hydrogen. The cytotoxicity profile of each particle was examined using a water-soluble tetrazolium-8 (WST-8) assay. At different time-points, a WST-8 assay, alkaline phosphatase (ALP) activity assay and alizarin red S (ARS) staining were used to determine the effects of rGO NPs on proliferation, differentiation and mineralization, respectively. The results suggest that graphene-based materials have potential as a platform for stem cells culture and biomedical applications.


Cell Differentiation/drug effects , Graphite/chemistry , Graphite/pharmacology , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Cells, Cultured , Humans , Mesenchymal Stem Cells/cytology , Oxidation-Reduction , Oxides/chemistry , Oxides/pharmacology
5.
Acta Crystallogr Sect E Struct Rep Online ; 70(Pt 9): o921, 2014 Sep 01.
Article En | MEDLINE | ID: mdl-25309253

In the title mol-ecule, C14H13NO2, the dihedral angle between the planes of the benzene rings is 65.18 (4)°. The central amide group has about the same degree of twist with respect to both ring planes, as indicated by the dihedral angles of 34.70 (8) and 30.62 (8)° between its plane and that of the phenyl and 4-meth-oxy-benzene rings, respectively. The C atom of the meth-oxy group is close to being coplanar with its attached ring [deviation = -0.112 (2) Å]. In the crystal, mol-ecules are linked by inter-amide N-H⋯O hydrogen bonds, which generate C(4) chains propagating in the [100] direction. Adajcent mol-ecules in the chain are related by translational symmetry.

6.
Biomater Res ; 18: 14, 2014.
Article En | MEDLINE | ID: mdl-26331065

BACKGROUND: M13 bacteriophages can be readily fabricated as nanofibers due to non-toxic bacterial virus with a nanofiber-like shape. In the present study, we prepared hybrid nanofiber matrices composed of poly(lactic-co-glycolic acid, PLGA) and M13 bacteriophages which were genetically modified to display the RGD peptide on their surface (RGD-M13 phage). RESULTS: The surface morphology and chemical composition of hybrid nanofiber matrices were characterized by scanning electron microscopy (SEM) and Raman spectroscopy, respectively. Immunofluorescence staining was conducted to investigate the existence of M13 bacteriophages in RGD-M13 phage/PLGA hybrid nanofibers. In addition, the attachment and proliferation of three different types of fibroblasts on RGD-M13 phage/PLGA nanofiber matrices were evaluated to explore how fibroblasts interact with these matrices. SEM images showed that RGD-M13 phage/PLGA hybrid matrices had the non-woven porous structure, quite similar to that of natural extracellular matrices, having an average fiber diameter of about 190 nm. Immunofluorescence images and Raman spectra revealed that RGD-M13 phages were homogeneously distributed in entire matrices. Moreover, the attachment and proliferation of fibroblasts cultured on RGD-M13 phage/PLGA matrices were significantly enhanced due to enriched RGD moieties on hybrid matrices. CONCLUSIONS: These results suggest that RGD-M13 phage/PLGA matrices can be efficiently used as biomimetic scaffolds for tissue engineering applications.

7.
J Nanosci Nanotechnol ; 14(11): 8458-63, 2014 Nov.
Article En | MEDLINE | ID: mdl-25958546

In this study, hyaluronic acid (HA)/poly(lactic-co-glycolic acid, PLGA) core/shell fiber meshes loaded with epigallocatechin-3-O-gallate (EGCG) (HA/PLGA-E) for application to tissue engineering scaffolds for skin regeneration were prepared via coaxial electrospinning. Physicochemical properties of HA/PLGA-E core/shell fiber meshes were characterized by SEM, Raman spectroscopy, contact angle, EGCG release profiling and in vitro degradation. Biomechanical properties of HA/PLGA-E meshes were also investigated by a tensile strength test. SEM images showed that HA/PLGA-E fiber meshes had a three-dimensional interconnected pore structure with an average fiber diameter of about 1270 nm. Raman spectra revealed that EGCG was uniformly dispersed in the PLGA shell of meshes. HA/PLGA-E meshes showed sustained EGCG release patterns by controlled diffusion and PLGA degradation over 4 weeks. EGCG loading did not adversely affect the tensile strength and elastic modulus of HA/PLGA meshes, while increased their hydrophilicity and surface energy. Attachment of human dermal fibroblasts on HA/PLGA-E meshes was appreciably increased and their proliferation was steadily retained during the culture period. These results suggest that HA/PLGA-E core/shell fiber meshes can be potentially used as scaffolds supporting skin regeneration.


Catechin/analogs & derivatives , Hyaluronic Acid/chemistry , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Skin/cytology , Tissue Scaffolds/chemistry , Catechin/chemistry , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fibroblasts/cytology , Humans , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Tissue Engineering
8.
Acta Haematol ; 131(1): 59-69, 2014.
Article En | MEDLINE | ID: mdl-24052005

BACKGROUND/AIMS: Constitutive activation of the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin signaling pathway preferentially occurs in aggressive blastoid variants of mantle cell lymphoma (MCL) and is implicated in the pathogenesis of this disease. In this study, we investigated the role of PI3K isoforms on proliferation of aggressive MCL cells. METHODS: The changes in cell viability, cell cycle distribution and apoptosis induction by the PI3K isoform-selective inhibitors were evaluated. The molecular basis underlying the effects of the specific inhibition of PI3K isoforms was investigated by Western blot analysis. RESULTS: Our results demonstrated that a class IA PI3K isoform is most commonly involved in the constitutive activation of Akt in aggressive MCL. Treatment with a p110α isoform-specific inhibitor induced prominent cell cycle arrest followed by apoptosis through complete abolishment of phosphorylated (p)-Akt and its downstream targets. An inhibitor of isoform p110δ induced moderate cell cycle arrest with downregulation of p-Akt and p-S6K. A dual inhibitor of p110α and p110δ GDC-0941 caused more prominent cell growth inhibition compared to selective p110α or p110δ inhibitors. Inhibition of the class IB PI3K isoform p110γ did not cause cell cycle arrest or induce apoptosis in MCL cells. CONCLUSION: These findings suggest that the therapeutic ablation of class IA PI3K may be a promising strategy for the treatment of refractory, aggressive MCL.


Lymphoma, Mantle-Cell/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Cell Cycle/drug effects , Cell Proliferation/drug effects , Chromones/pharmacology , Humans , Indazoles , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Morpholines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidinones/pharmacology , Sulfonamides
9.
J Mol Med (Berl) ; 91(12): 1383-97, 2013 Dec.
Article En | MEDLINE | ID: mdl-23955073

UNLABELLED: Both phosphatidylinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin signaling and antiapoptotic Bcl-2 family members are critical for survival of acute myeloid leukemia (AML) cells. Here, we demonstrate the antileukemic effects of simultaneous inhibition of PI3K by the selective class I PI3K inhibitor GDC-0941 and of Bcl-2 family members by the BH3 mimetic ABT-737 in the context of the bone marrow microenvironment, where hypoxia and interactions with bone marrow stromal cells promote AML cell survival and chemoresistance. The combination of GDC-0941 and ABT-737 profoundly downregulated antiapoptotic Mcl-1 expression levels, activated BAX, and induced mitochondrial apoptosis in AML cells co-cultured with bone marrow stromal cells under hypoxic conditions. Hypoxia caused degradation of Mcl-1 and rendered Mcl-1-overexpressing OCI-AML3 cells sensitive to ABT-737. Our findings suggest that pharmacologic PI3K inhibition by GDC-0941 enhances ABT-737-induced leukemia cell death even under the protective conditions afforded by the bone marrow microenvironment. KEY MESSAGE: Combined blockade of PI3K and Bcl-2 pathways down-regulates anti-apoptotic Mcl-1 expression PI3K and Bcl-2 induced Mcl-1 down-regulation activates BAX PI3K and Bcl-2 blockage induces apoptosis in AML under hypoxic BM microenvironment.


Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Bone Marrow/drug effects , Bone Marrow/metabolism , Indazoles/pharmacology , Leukemia, Myeloid, Acute/metabolism , Nitrophenols/pharmacology , Sulfonamides/pharmacology , Tumor Microenvironment/drug effects , Bone Marrow/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Drug Synergism , Gene Expression Regulation, Leukemic , Humans , Hypoxia/metabolism , Leukemia, Myeloid, Acute/genetics , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Phosphoinositide-3 Kinase Inhibitors , Piperazines/pharmacology , RNA Interference , Tumor Microenvironment/genetics
10.
PLoS One ; 8(6): e62785, 2013.
Article En | MEDLINE | ID: mdl-23826077

Hypoxia and interactions with bone marrow (BM) stromal cells have emerged as essential components of the leukemic BM microenvironment in promoting leukemia cell survival and chemoresistance. High levels of transforming growth factor beta 1 (TGFß1) produced by BM stromal cells in the BM niche regulate cell proliferation, survival, and apoptosis, depending on the cellular context. Exogenous TGFß1 induced accumulation of acute myeloid leukemia (AML) cells in a quiescent G0 state, which was further facilitated by the co-culture with BM-derived mesenchymal stem cells (MSCs). In turn, TGFß-neutralizing antibody 1D11 abrogated rhTGFß1 induced cell cycle arrest. Blocking TGFß with 1D11 further enhanced cytarabine (Ara-C)-induced apoptosis of AML cells in hypoxic and in normoxic conditions. Additional constituents of BM niche, the stroma-secreted chemokine CXCL12 and its receptor CXCR4 play crucial roles in cell migration and stroma/leukemia cell interactions. Treatment with 1D11 combined with CXCR4 antagonist plerixafor and Ara-C decreased leukemia burden and prolonged survival in an in vivo leukemia model. These results indicate that blockade of TGFß by 1D11 and abrogation of CXCL12/CXCR4 signaling may enhance the efficacy of chemotherapy against AML cells in the hypoxic BM microenvironment.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Marrow/drug effects , Cytarabine/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/immunology , Tumor Microenvironment/drug effects , Animals , Antibodies, Neutralizing/metabolism , Benzylamines , Bone Marrow/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chemokine CXCL12/metabolism , Coculture Techniques , Cyclams , Female , Heterocyclic Compounds/pharmacology , Humans , Leukemia, Myeloid, Acute/metabolism , Mice, SCID , Neoplasm Transplantation , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Tumor Microenvironment/physiology
11.
Cancer Lett ; 329(1): 45-58, 2013 Feb 01.
Article En | MEDLINE | ID: mdl-23036488

We investigated the antileukemia effects and molecular mechanisms of apoptosis induction by simultaneous blockade of PI3K and mutant FLT3 in AML cells grown under hypoxia in co-cultures with bone marrow stromal cells. Combined treatment with selective class I PI3K inhibitor GDC-0941 and sorafenib reversed the protective effects of bone marrow stromal cells on FLT3-mutant AML cells in hypoxia, which was associated with downregulation of Pim-1 and Mcl-1 expression levels. These findings suggest that combined inhibition of PI3K and FLT3-ITD may constitute a targeted approach to eradicating chemoresistant AML cells sequestered in hypoxic bone marrow niches.


Antineoplastic Agents/pharmacology , Bone Marrow/drug effects , Indazoles/pharmacology , Leukemia, Myeloid, Acute/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Sulfonamides/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Apoptosis/drug effects , Bone Marrow/metabolism , Cell Hypoxia/drug effects , Cellular Microenvironment/drug effects , Coculture Techniques , Enzyme Inhibitors/pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mutation , Myeloid Cell Leukemia Sequence 1 Protein , Niacinamide/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Sorafenib , Stromal Cells/drug effects , Stromal Cells/metabolism , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
12.
Cancer Lett ; 299(2): 161-70, 2010 Dec 28.
Article En | MEDLINE | ID: mdl-20850924

This study demonstrated a pronounced synergistic growth-inhibitory effect of an MDM2 inhibitor Nutlin-3 and a proteasome inhibitor bortezomib in mantle cell lymphoma (MCL) cells regardless of TP53 mutant status and innate bortezomib sensitivity. In the mutant TP53 MCL cells which are intrinsically resistant to bortezomib, the combination of Nutlin-3/bortezomib synergistically induced cytotoxicity through the mitochondrial apoptotic pathway mediated by transcription-independent upregulation of NOXA, sequestration of MCL-1, activation of BAX, BAK, caspase-9 and -3. In the bortezomib sensitive wild-type TP53 MCL cells, the Nutlin-3/bortezomib combination caused G0/G1 cell cycle arrest followed by the increase in apoptosis induction. These findings indicate potential therapeutic efficacy of Nutlin-3/bortezomib combination for the treatment of chemorefractory MCL.


Apoptosis/drug effects , Boronic Acids/pharmacology , Imidazoles/pharmacology , Piperazines/pharmacology , Pyrazines/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Bortezomib , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Humans , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Mutation , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
13.
Clin Cancer Res ; 15(3): 933-42, 2009 Feb 01.
Article En | MEDLINE | ID: mdl-19188164

PURPOSE: Mantle cell lymphoma (MCL) has one of the poorest prognoses of the non-Hodgkin's lymphomas, and novel therapeutic approaches are needed. We wished to determine whether Nutlin-3, a novel small-molecule murine double minute 2 (MDM2) antagonist that efficiently activates TP53, might be effective in inducing cell death in MCL. EXPERIMENTAL DESIGN: MCL cell lines with known TP53 status were treated with Nutlin-3, and biological and biochemical consequences were studied. Synergies with the prototypic genotoxic agent doxorubicin and the novel proteasome inhibitor bortezomib were assessed. RESULTS: Nutlin-3 resulted in a reduction in cell proliferation/viability (IC50 < 10 micromol/L), an increase in the apoptotic fraction, and cell cycle arrest in wild-type (wt) TP53 Z-138 and Granta 519 cells. These effects were accompanied by TP53 accumulation and induction of TP53-dependent proteins p21, MDM2, Puma, and Noxa. Cell cycle arrest was characterized by suppression of S phase and an increase in the G0-G1 and G2-M fractions and accompanied by suppression of total and phosphorylated retinoblastoma protein and a decrease in G2-M-associated proteins cyclin B and CDC2. The combination of Nutlin-3 with doxorubicin or bortezomib was synergistic in wt-TP53 MCL cells. Nutlin-3 also induced cell cycle arrest and reduced cell viability in the mutant TP53 MINO cells but at a significantly higher IC50 (22.5 micromol/L). These effects were associated with induction of the TP53 homologue p73, slight increases in p21 and Noxa, and caspase activation. Nutlin-3 and bortezomib synergistically inhibited cell growth of MINO. CONCLUSION: These findings suggest that the MDM2 antagonist Nutlin-3 may be an effective agent in the treatment of MCL with or without wt-TP53.


Imidazoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Boronic Acids/administration & dosage , Bortezomib , Cell Cycle/drug effects , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/administration & dosage , Humans , Imidazoles/administration & dosage , Lymphoma, Mantle-Cell/pathology , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Piperazines/administration & dosage , Proto-Oncogene Proteins/metabolism , Pyrazines/administration & dosage , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
14.
Br J Haematol ; 142(2): 192-201, 2008 Jun.
Article En | MEDLINE | ID: mdl-18492113

Transforming growth factor beta1 (TGF-beta1) is an essential regulator of cell proliferation, survival and apoptosis, depending on the cellular context. TGF-beta1 is also known to affect cell-to-cell interactions between tumour cells and stromal cells. We investigated the role of TGF-beta1 in the survival of myelo-monocytic leukaemia cell lines co-cultured with bone marrow (BM)-derived mesenchymal stem cells (MSC). Treatment with recombinant human (rh)TGF-beta1 inhibited spontaneous and cytarabine-induced apoptosis in U937 cells, most prominently in U937 cells directly attached to MSCs. Conversely, the pro-survival effects of TGF-beta1 were inhibited by LY2109761 or TGF-beta1 neutralizing antibody. rhTGF-beta1 increased pro-survival phosphorylation of Akt, which was inhibited by LY2109761. The combination of rhTGF-beta1 and MSC co-culture induced significant upregulation of C/EBPbeta gene (CEBPB) and protein expression along with increased C/EBPbeta liver-enriched activating protein: liver-enriched inhibitory protein ratio, suggesting the novel role of C/EBPbeta in TGF-beta1-mediated U937 cell survival in the context of stromal cell support. In summary, these results indicate that TGF-beta1 produced by BM stromal cells promotes the survival and chemoresistance of leukaemia cells under the direct cell-to-cell interactions. The blockade of TGF-beta signalling by LY2109761, which effectively inhibited the pro-survival signalling, may enhance the efficacy of chemotherapy against myelo-monocytic leukaemic cells in the BM microenvironment.


Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Leukemia/drug therapy , Pyrazoles/pharmacology , Pyrroles/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta1/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor/metabolism , Cells, Cultured/drug effects , Coculture Techniques , Humans , Leukemia/metabolism , Lipopolysaccharide Receptors/metabolism , Mesenchymal Stem Cells/metabolism , Stromal Cells/metabolism , Transforming Growth Factor beta1/metabolism , U937 Cells/metabolism
15.
Mol Cancer Ther ; 7(1): 48-58, 2008 Jan.
Article En | MEDLINE | ID: mdl-18202009

Chronic myelogenous leukemia (CML) is driven by constitutively activated Bcr-Abl tyrosine kinase, which causes the defective adhesion of CML cells to bone marrow stroma. The overexpression of p210Bcr-Abl was reported to down-regulate CXCR4 expression, and this is associated with the cell migration defects in CML. We proposed that tyrosine kinase inhibitors, imatinib or INNO-406, may restore CXCR4 expression and cause the migration of CML cells to bone marrow microenvironment niches, which in turn results in acquisition of stroma-mediated chemoresistance of CML progenitor cells. In KBM5 and K562 cells, imatinib, INNO-406, or IFN-alpha increased CXCR4 expression and migration. This increase in CXCR4 levels on CML progenitor cells was likewise found in samples from CML patients treated with imatinib or IFN-alpha. Imatinib induced G0-G1 cell cycle block in CML cells, which was further enhanced in a mesenchymal stem cell (MSC) coculture system. MSC coculture protected KBM-5 cells from imatinib-induced cell death. These antiapoptotic effects were abrogated by the CXCR4 antagonist AMD3465 or by inhibitor of integrin-linked kinase QLT0267. Altogether, these findings suggest that the up-regulation of CXCR4 by imatinib promotes migration of CML cells to bone marrow stroma, causing the G0-G1 cell cycle arrest and hence ensuring the survival of quiescent CML progenitor cells.


Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Movement/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptors, CXCR4/metabolism , Antigens, CD34/metabolism , Benzamides , Cell Line, Tumor , Cell Survival/drug effects , Humans , Imatinib Mesylate , Interferon-alpha/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Up-Regulation/drug effects
16.
Cancer Res ; 67(2): 684-94, 2007 Jan 15.
Article En | MEDLINE | ID: mdl-17234779

Integrin-linked kinase (ILK) directly interacts with beta integrins and phosphorylates Akt in a phosphatidylinositol 3-kinase (PI3K)-dependent manner. In this study, we examined the functional role of ILK activation in leukemic and bone marrow stromal cells on their direct contact. Coculture of leukemic NB4 cells with bone marrow-derived stromal mesenchymal stem cells (MSC) resulted in robust activation of multiple signaling pathways, including ILK/Akt, extracellular signal-regulated kinase 1/2 (ERK1/2), signal transducers and activators of transcription 3 (STAT3), and Notch1/Hes. Blockade of PI3K or ILK signaling with pharmacologic inhibitors LY294002 or QLT0267 specifically inhibited stroma-induced phosphorylation of Akt and glycogen synthase kinase 3beta, suppressed STAT3 and ERK1/2 activation, and decreased Notch1 and Hes1 expression in leukemic cells. This resulted in induction of apoptosis in both leukemic cell lines and in primary acute myelogenous leukemia samples that was not abrogated by MSC coculture. In turn, leukemic cells growing in direct contact with bone marrow stromal elements induce activation of Akt, ERK1/2, and STAT3 signaling in MSC, accompanied by significant increase in Hes1 and Bcl-2 proteins, which were all suppressed by QLT0267 and LY294002. In summary, our results indicate reciprocal activation of ILK/Akt in both leukemic and bone marrow stromal cells. We propose that ILK/Akt is a proximal signaling pathway critical for survival of leukemic cells within the bone marrow microenvironment. Hence, disruption of these interactions by ILK inhibitors represents a potential novel therapeutic strategy to eradicate leukemia in the bone marrow microenvironment by simultaneous targeting of both leukemic cells and activated bone marrow stromal cells.


Cell Communication/physiology , Leukemia, Promyelocytic, Acute/enzymology , Mesenchymal Stem Cells/enzymology , Protein Serine-Threonine Kinases/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Cell Survival/physiology , Chromones/pharmacology , Coculture Techniques , Enzyme Activation/drug effects , Homeodomain Proteins/metabolism , Humans , Janus Kinases/metabolism , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Mesenchymal Stem Cells/cytology , Mitogen-Activated Protein Kinase Kinases/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Notch/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Stromal Cells/cytology , Stromal Cells/enzymology , Transcription Factor HES-1
17.
Rinsho Ketsueki ; 47(10): 1364-71, 2006 Oct.
Article Ja | MEDLINE | ID: mdl-17094575

The goal of this study was to elucidate the functional roles of PI3K/AKT and MEK/ERK signaling on the proliferation and apoptosis of STI571-sensitive and -resistant CML cell lines in a co-culture system with human marrow stromal cells (MSCs), mimicking the bone marrow microenvironment. The phosphorylation of AKT and ERK was enhanced by co-culture with MSCs in both STI571-sensitive KBM-5 and STI571-resistant KBM-5/STI cells. In KBM-5 cells, the STI571 and PI3K inhibitor LY294002 combination was effective on apoptosis induction in the MSC co-culture system. In KBM-5/STI cells, treatment with LY294002 or PD98059 alone resulted in massive apoptosis, which was enhanced by co-culture with MSCs. These results provide a rationale for multi-molecular target therapy approaches based on a combination of signal transduction inhibitors with STI571 in CML.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Marrow Cells/cytology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Phosphoinositide-3 Kinase Inhibitors , Piperazines/pharmacology , Pyrimidines/pharmacology , Stromal Cells/drug effects , Benzamides , Cell Proliferation , Chromones/pharmacology , Coculture Techniques , Flavonoids/pharmacology , Humans , Imatinib Mesylate , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation
18.
Ann Hematol ; 85(10): 689-704, 2006 Oct.
Article En | MEDLINE | ID: mdl-16832676

The acute promyelocytic leukemia-specific PML-RARalpha fusion protein is a dominant-negative transcriptional repressor of retinoic acid receptor (RAR) target genes, which recruits HDAC and corepressor proteins and inhibits coactivators. Another oncogenic transcription factor, AML1-ETO, was proposed to cause an HDAC-dependent repression of RAR target genes. The RAR target RARbeta2 gene has been reported to be frequently silenced by hypermethylation in many types of cancer cells. We examined the methylation status of the RARbeta2 and asked if demethylation could reverse ATRA resistance in ATRA-resistant PML-RARalpha and AML1-ETO-positive cells. PML-RARalpha positive NB4 and its ATRA-resistant subvariant MR2 and AML1-ETO expressing Kasumi-1 cells had heterozygous methylation of RARbeta2. Although DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine partially reversed RARbeta2 CpG methylation in these cells, it did not significantly enhance ATRA-induced RARbeta2 mRNA expression and induction of maturation. However, the histone acetylase inhibitor SAHA combined with ATRA significantly reactivated RARbeta2 mRNA both in NB4 and MR2 cells with degradation of PML-RARalpha, which was associated with maturation. In contrast, SAHA did not affect AML1-ETO levels and failed to induce RARbeta2 expression and maturation in Kasumi-1 cells. In primary AML samples, RARbeta2 expression was uniformly low; however, no specific correlation was observed between the methylation of the RARbeta2 gene and expression of the fusion proteins, PML-RARalpha, and AML1-ETO. These results demonstrate that oncogenic PML-RARalpha and AML1-ETO translocations are rarely associated with RARbeta2 promoter methylation in primary AML samples.


Core Binding Factor Alpha 2 Subunit/biosynthesis , DNA Methylation , Leukemia, Promyelocytic, Acute/metabolism , Oncogene Proteins, Fusion/biosynthesis , Promoter Regions, Genetic , Receptors, Retinoic Acid/biosynthesis , Translocation, Genetic , Antineoplastic Agents/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Core Binding Factor Alpha 2 Subunit/genetics , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , Decitabine , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Histone Deacetylases/biosynthesis , Histone Deacetylases/genetics , Humans , K562 Cells , Leukemia, Promyelocytic, Acute/genetics , Oncogene Proteins, Fusion/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RUNX1 Translocation Partner 1 Protein , Receptors, Retinoic Acid/genetics , Translocation, Genetic/drug effects , Translocation, Genetic/genetics , Tretinoin/pharmacology , U937 Cells
19.
Blood ; 107(4): 1546-54, 2006 Feb 15.
Article En | MEDLINE | ID: mdl-16223781

The multidrug resistance 1 (MDR1) gene product P-glycoprotein (P-gp) is frequently implicated in cross-resistance of tumors to chemotherapeutic drugs. In contrast, acute promyelocytic leukemia (APL) cells do not express MDR1 and are highly sensitive to anthracyclines. The combination of ATRA and the novel histone deacetylase inhibitor (HDACI) depsipeptide (FK228) induced P-gp expression and prevented growth inhibition and apoptosis in NB4 APL cells subsequently exposed to doxorubicin (DOX). ATRA/FK228 treatment after exposure to DOX, however, enhanced apoptosis. Both agents, ATRA or FK228, induced MDR1 mRNA. This effect was significantly enhanced by ATRA/FK228 administered in combination, due in part to increased H4 and H3-Lys9 acetylation of the MDR1 promoter and recruitment of the nuclear transcription factor Y alpha (NFYA) transcription activator to the CCAAT box. Cotreatment with specific P-gp inhibitor PSC833 reversed cytoprotective effects of ATRA/FK228. G1 cell-cycle arrest and p21 mRNA induction were also observed in response to ATRA/FK228, which may restrict DOX-induced apoptosis of cells in G2 phase. These results indicate that epigenetic mechanisms involving NF-YA transcription factor recruitment and histone acetylation are activated by ATRA and HDACI, induce MDR1 in APL cells, and point to the critical importance of mechanism-based sequential therapy in future clinical trials that combine HDAC inhibitors, ATRA, and anthracyclines.


ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antibiotics, Antineoplastic/pharmacology , Depsipeptides/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/physiology , Leukemia, Promyelocytic, Acute/genetics , Cell Cycle/drug effects , Enzyme Inhibitors/pharmacology , Genes, MDR , Histone Deacetylase Inhibitors , Humans
...