Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 9(11)2021 11.
Article in English | MEDLINE | ID: mdl-34824159

ABSTRACT

BACKGROUND: Natural killer (NK) cells require a functional lytic granule machinery to mediate effective antitumor responses. Evading the lytic cargo deployed at the immune synapse (IS) could be a critical step for cancer progression through yet unidentified mechanisms. METHODS: NK cell antibody-dependent cellular cytotoxicity (ADCC) is a major determinant of the clinical efficacy of some therapeutic antibodies including the anti-HER2 Trastuzumab. Thus, we screened sera of Trastuzumab-resistant HER2 +patients with breast cancer for molecules that could inhibit NK cell ADCC. We validated our findings in vitro using cytotoxicity assays and confocal imaging of the lytic granule machinery and in vivo using syngeneic and xenograft murine models. RESULTS: We found that sera from Trastuzumab-refractory patients could inhibit healthy NK cell ADCC in vitro. These sera contained high levels of the inflammatory protein chitinase 3-like 1 (CHI3L1) compared with sera from responders and healthy controls. We demonstrate that recombinant CHI3L1 inhibits both ADCC and innate NK cell cytotoxicity. Mechanistically, CHI3L1 prevents the correct polarization of the microtubule-organizing center along with the lytic granules to the IS by hindering the receptor of advanced glycation end-products and its downstream JNK signaling. In vivo, CHI3L1 administration drastically impairs the control of NK cell-sensitive tumors, while CHI3L1 blockade synergizes with ADCC to cure mice with HER2 +xenografts. CONCLUSION: Our work highlights a new paradigm of tumor immune escape mediated by CHI3L1 which acts on the cytotoxic machinery and prevents granule polarization. Targeting CHI3L1 could mitigate immune escape and potentiate antibody and cell-based immunotherapies.


Subject(s)
Chitinase-3-Like Protein 1/metabolism , Immune Evasion/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasms/genetics , Animals , Female , Humans , Mice
3.
MAbs ; 11(8): 1402-1414, 2019.
Article in English | MEDLINE | ID: mdl-31526159

ABSTRACT

High specificity accompanied with the ability to recruit immune cells has made recombinant therapeutic antibodies an integral part of drug development. Here we present a generic approach to generate two novel IgG-derived antibody formats that are based on a modification of the CrossMab technology. MoAbs harbor two heavy chains (HCs) resulting in one binding entity and one fragment crystallizable region (Fc), whereas DuoMabs are composed of four HCs harboring two binding entities and two Fc regions linked at a disulfide-bridged hinge. The latter bivalent format is characterized by avidity-enhanced target cell binding while simultaneously increasing the 'Fc-load' on the surface. DuoMabs were shown to be producible in high yield and purity and bind to surface cells with affinities comparable to IgGs. The increased Fc load directed at the surface of target cells by DuoMabs modulates their antibody-dependent cell-mediated cytotoxicity competency toward target cells, making them attractive for applications that require or are modulated by FcR interactions.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Antibodies, Bispecific/chemistry , Antibodies, Monoclonal/chemistry , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry
4.
Clin Cancer Res ; 25(19): 5890-5900, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31285373

ABSTRACT

PURPOSE: Genetically engineered T cells are powerful anticancer treatments but are limited by safety and specificity issues. We herein describe an MHC-unrestricted modular platform combining autologous T cells, transduced with a targetable synthetic agonistic receptor (SAR), with bispecific antibodies (BiAb) that specifically recruit and activate T cells for tumor killing. EXPERIMENTAL DESIGN: BiAbs of different formats were generated by recombinant expression. T cells were retrovirally transduced with SARs. T-cell activation, proliferation, differentiation, and T-cell-induced lysis were characterized in three murine and human tumor models in vitro and in vivo. RESULTS: Murine T cells transduced with SAR composed of an extracellular domain EGFRvIII fused to CD28 and CD3ζ signaling domains could be specifically recruited toward murine tumor cells expressing EpCAM by anti-EGFRvIII × anti-EpCAM BiAb. BiAb induced selective antigen-dependent activation, proliferation of SAR T cells, and redirected tumor cell lysis. Selectivity was dependent on the monovalency of the antibody for EGFRvIII. We identified FAS ligand as a major mediator of killing utilized by the T cells. Similarly, human SAR T cells could be specifically redirected toward mesothelin-expressing human pancreatic cancer cells. In vivo, treatment with SAR T cells and BiAb mediated antitumoral activity in three human pancreatic cancer cell xenograft models. Importantly, SAR activity, unlike CAR activity, was reversible in vitro and in vivo. CONCLUSIONS: We describe a novel ACT platform with antitumor activity in murine and human tumor models with a distinct mode of action that combines adoptive T-cell therapy with bispecific antibodies.


Subject(s)
Antibodies, Bispecific/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , ErbB Receptors/immunology , Immunotherapy, Adoptive/methods , Pancreatic Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/genetics , Epithelial Cell Adhesion Molecule/immunology , Epithelial Cell Adhesion Molecule/metabolism , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mesothelin , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Pancreatic Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Protein Eng Des Sel ; 29(10): 467-475, 2016 10.
Article in English | MEDLINE | ID: mdl-27578890

ABSTRACT

Monoclonal antibody-based targeted tumor therapy has greatly improved treatment options for patients. Antibodies against oncogenic receptor tyrosine kinases (RTKs), especially the ErbB receptor family, are prominent examples. However, long-term efficacy of such antibodies is limited by resistance mechanisms. Tumor evasion by a priori or acquired activation of other kinases is often causative for this phenomenon. These findings led to an increasing number of combination approaches either within a protein family, e.g. the ErbB family or by targeting RTKs of different phylogenetic origin like HER1 and cMet or HER1 and IGF1R. Progress in antibody engineering technology enabled generation of clinical grade bispecific antibodies (BsAbs) to design drugs inherently addressing such resistance mechanisms. Limited data are available on multi-specific antibodies targeting three or more RTKs. In the present study, we have evaluated the cloning, eukaryotic expression and purification of tetraspecific, tetravalent Fc-containing antibodies targeting HER3, cMet, HER1 and IGF1R. The antibodies are based on the combination of single-chain Fab and Fv fragments in an IgG1 antibody format enhanced by the knob-into-hole technology. They are non-agonistic and inhibit tumor cell growth comparable to the combination of four parental antibodies. Importantly, TetraMabs show improved apoptosis induction and tumor growth inhibition over individual monospecific or BsAbs in cellular assays. In addition, a mimicry assay to reflect heterogeneous expression of antigens in a tumor mass was established. With this novel in vitro assay, we can demonstrate the superiority of a tetraspecific antibody to bispecific tumor antigen-binding antibodies in early pre-clinical development.


Subject(s)
Molecular Targeted Therapy/methods , Receptor Protein-Tyrosine Kinases/immunology , Single-Chain Antibodies/immunology , Antibody Specificity , Apoptosis/immunology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Enzyme Activation , Humans , Protein Engineering , Receptor Protein-Tyrosine Kinases/metabolism , Single-Chain Antibodies/genetics
6.
Immunol Rev ; 270(1): 165-77, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26864111

ABSTRACT

Hapten-binding antibodies have for more than 50 years played a pivotal role in immunology, paving the way to antibody generation (as haptens are very important and robust immunogens), to antibody characterization (as the first structures generated more than 40 years ago were those of hapten binders), and enabled and expanded antibody engineering technologies. The latter field of engineered antibodies evolved over many years and many steps resulting in recombinant humanized or human-derived antibody derivatives in multiple formats. Today, hapten-binding antibodies are applied not only as reagents and tools (where they still play an important part) but evolved also to engineered targeting and pretargeting vehicles for disease diagnosis and therapy. Here we describe recent applications of hapten-binding antibodies and of engineered mono- and bispecific hapten-binding antibody derivatives. We have designed and applied these molecules for the modulation of the pharmacokinetic properties of small compounds or peptides. They are also integrated as additional binding entities into bispecific antibody formats. Here they serve as non-covalent or covalent coupling modules to haptenylated compounds, to enable targeted payload delivery to disease tissues or cells.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , Drug Delivery Systems , Haptens/immunology , Haptens/metabolism , Molecular Targeted Therapy , Protein Engineering , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/pharmacology , Antigens, Surface/immunology , Antigens, Surface/metabolism , Biomarkers , Haptens/chemistry , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Protein Binding , Recombinant Fusion Proteins
7.
J Natl Cancer Inst ; 107(1): 364, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25424197

ABSTRACT

BACKGROUND: One bottleneck for adoptive T cell therapy (ACT) is recruitment of T cells into tumors. We hypothesized that combining tumor-specific T cells, modified with a marker antigen and a bispecific antibody (BiAb) that selectively recognizes transduced T cells and tumor cells would improve T cell recruitment to tumors and enhance therapeutic efficacy. METHODS: SV40 T antigen-specific T cells from T cell receptor (TCR)-I-transgenic mice were transduced with a truncated human epidermal growth factor receptor (EGFR) as a marker protein. Targeting and killing by combined ACT and anti-EGFR-anti-EpCAM BiAb therapy was analyzed in C57Bl/6 mice (n = six to 12 per group) carrying subcutaneous tumors of the murine gastric cancer cell line GC8 (SV40(+) and EpCAM(+)). Anti-EGFR x anti-c-Met BiAb was used for targeting of human tumor-specific T cells to c-Met(+) human tumor cell lines. Differences between experimental conditions were analyzed using the Student's t test, and differences in tumor growth with two-way analysis of variance. Overall survival was analyzed by log-rank test. All statistical tests were two-sided. RESULTS: The BiAb linked EGFR-transduced T cells to tumor cells and enhanced tumor cell lysis. In vivo, the combination of ACT and Biab produced increased T cell infiltration of tumors, retarded tumor growth, and prolonged survival compared with ACT with a control antibody (median survival 95 vs 75 days, P < .001). In human cells, this strategy enhanced recruitment of human EGFR-transduced T cells to immobilized c-Met and recognition of tyrosinase(+) melanoma cells by TCR-, as well as of CEA(+) colon cancer cells by chimeric antigen receptor (CAR)-modified T cells. CONCLUSIONS: BiAb recruitment of tumor-specific T cells transduced with a marker antigen to tumor cells may enhance efficacy of ACT.


Subject(s)
Adoptive Transfer , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , ErbB Receptors/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptors, Antigen, T-Cell/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/therapy , T-Lymphocytes/immunology , Analysis of Variance , Animals , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , ErbB Receptors/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-met/immunology , Transduction, Genetic
8.
Nat Med ; 18(10): 1550-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23001182

ABSTRACT

We found that hematopoietic cell-specific Lyn substrate 1 (HCLS1 or HS1) is highly expressed in human myeloid cells and that stimulation with granulocyte colony-stimulating factor (G-CSF) leads to HCLS1 phosphorylation. HCLS1 binds the transcription factor lymphoid-enhancer binding factor 1 (LEF-1), transporting LEF-1 into the nucleus upon G-CSF stimulation and inducing LEF-1 autoregulation. In patients with severe congenital neutropenia, inherited mutations in the gene encoding HCLS1-associated protein X-1 (HAX1) lead to profound defects in G-CSF-triggered phosphorylation of HCLS1 and subsequently to reduced autoregulation and expression of LEF-1. Consistent with these results, HCLS1-deficient mice are neutropenic. In bone marrow biopsies of the majority of tested patients with acute myeloid leukemia, HCLS1 protein expression is substantially elevated, associated with high levels of G-CSF synthesis and, in some individuals, a four-residue insertion in a proline-rich region of HCLS1 protein known to accelerate intracellular signaling. These data demonstrate the importance of HCLS1 in myelopoiesis in vitro and in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Blood Proteins/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Myelopoiesis , Neutropenia/congenital , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing/metabolism , Animals , Base Sequence , Blood Proteins/genetics , Cell Line , Cell Proliferation , Congenital Bone Marrow Failure Syndromes , Female , Granulocytes/metabolism , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Myelopoiesis/genetics , Neutropenia/genetics , Neutropenia/physiopathology , Phosphorylation , RNA Interference , RNA, Small Interfering , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Sequence Analysis, DNA , Signal Transduction
9.
Protein Eng Des Sel ; 25(10): 571-80, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22976197

ABSTRACT

We have designed bispecific antibodies that bind one target (anti-Her3) in a bivalent IgG-like manner and contain one additional binding entity (anti-cMet) composed of one V(H) and one V(L) domain connected by a disulfide bond. The molecules are assembled by fusing a V(H,Cys44) domain via flexible connector peptides to the C-terminus of one H-chain (heavy chain), and a V(L,Cys100) to another H-chain. To ensure heterodimerization during expression in mammalian cells, we introduced complementary knobs-into-holes mutations into the different H-chains. The IgG-shaped trivalent molecules carry as third binding entity one disulfide-stabilized Fv (dsFv) without a linker between V(H) and V(L). Tethering the V(H) and V(L) domains at the C-terminus of the C(H)3 domain decreases the on-rates of the dsFv to target antigens without affecting off-rates. Steric hindrance resolves upon removal of one side of the double connection by proteolysis: this improves flexibility and accessibility of the dsFv and fully restores antigen access and affinity. This technology has multiple applications: (i) in cases where single-chain linkers are not desired, dsFvs without linkers can be generated by addition of furin site(s) in the connector that are processed during expression within mammalian cells; (ii) highly active (toxic) entities which affect expression can be produced as inactive dsFvs and subsequently be activated (e.g. via PreScission cleavage) during purification; (iii) entities can be generated which are targeted by the unrestricted binding entity and can be activated by proteases in target tissues. For example, Her3-binding molecules containing linkers with recognition sequences for matrix metalloproteases or urokinase, whose inactivated cMet binding site is activated by proteolytic processing.


Subject(s)
Antibodies, Bispecific/chemistry , Antibodies, Bispecific/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Binding Sites, Antibody , Cell Line , Disulfides/chemistry , Gene Expression , Humans , Matrix Metalloproteinase 2/immunology , Matrix Metalloproteinase 9/immunology , Peptide Hydrolases/metabolism , Protein Engineering , Proteolysis , Receptor, ErbB-3/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Urokinase-Type Plasminogen Activator/immunology
10.
MAbs ; 4(6): 653-63, 2012.
Article in English | MEDLINE | ID: mdl-22925968

ABSTRACT

The development of bispecific antibodies has attracted substantial interest, and many different formats have been described. Those specifically containing an Fc part are mostly tetravalent, such as stabilized IgG-scFv fusions or dual-variable domain (DVD) IgGs. However, although they exhibit IgG-like properties and technical developability, these formats differ in size and geometry from classical IgG antibodies. Thus, considerable efforts focus on bispecific heterodimeric IgG antibodies that more closely mimic natural IgG molecules. The inherent chain association problem encountered when producing bispecific heterodimeric IgG antibodies can be overcome by several methods. While technologies like knobs-into-holes (KiH) combined with a common light chain or the CrossMab technology enforce the correct chain association, other approaches, e.g., the dual-acting Fab (DAF) IgGs, do not rely on a heterodimeric Fc part. This review discusses the state of the art in bispecific heterodimeric IgG antibodies, with an emphasis on recent progress.


Subject(s)
Antibodies, Bispecific/metabolism , Immunotherapy/methods , Single-Chain Antibodies/metabolism , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/therapeutic use , Antibody Affinity , Drug Design , Humans , Immunotherapy/trends , Protein Binding , Protein Engineering , Protein Multimerization , Single-Chain Antibodies/genetics , Single-Chain Antibodies/therapeutic use
11.
Proc Natl Acad Sci U S A ; 108(27): 11187-92, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21690412

ABSTRACT

We describe a generic approach to assemble correctly two heavy and two light chains, derived from two existing antibodies, to form human bivalent bispecific IgG antibodies without use of artificial linkers. Based on the knobs-into-holes technology that enables heterodimerization of the heavy chains, correct association of the light chains and their cognate heavy chains is achieved by exchange of heavy-chain and light-chain domains within the antigen binding fragment (Fab) of one half of the bispecific antibody. This "crossover" retains the antigen-binding affinity but makes the two arms so different that light-chain mispairing can no longer occur. Applying the three possible "CrossMab" formats, we generated bispecific antibodies against angiopoietin-2 (Ang-2) and vascular endothelial growth factor A (VEGF-A) and show that they can be produced by standard techniques, exhibit stabilities comparable to natural antibodies, and bind both targets simultaneously with unaltered affinity. Because of its superior side-product profile, the CrossMab(CH1-CL) was selected for in vivo profiling and showed potent antiangiogenic and antitumoral activity.


Subject(s)
Antibodies, Bispecific/biosynthesis , Antibodies, Bispecific/chemistry , Immunoglobulin G/biosynthesis , Immunoglobulin G/chemistry , Angiopoietin-2/immunology , Animals , Antibodies, Bispecific/metabolism , Antibody Affinity , Antibody Specificity , Cell Line , Cell Line, Tumor , Female , Humans , Immunoglobulin G/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Models, Molecular , Neovascularization, Physiologic , Protein Engineering , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Vascular Endothelial Growth Factor A/immunology
12.
Cell ; 133(1): 103-15, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18394993

ABSTRACT

RanBP2 is a nucleoporin with SUMO E3 ligase activity that functions in both nucleocytoplasmic transport and mitosis. However, the biological relevance of RanBP2 and the in vivo targets of its E3 ligase activity are unknown. Here we show that animals with low amounts of RanBP2 develop severe aneuploidy in the absence of overt transport defects. The main chromosome segregation defect in cells from these mice is anaphase-bridge formation. Topoisomerase IIalpha (Topo IIalpha), which decatenates sister centromeres prior to anaphase onset to prevent bridges, fails to accumulate at inner centromeres when RanBP2 levels are low. We find that RanBP2 sumoylates Topo IIalpha in mitosis and that this modification is required for its proper localization to inner centromeres. Furthermore, mice with low amounts of RanBP2 are highly sensitive to tumor formation. Together, these data identify RanBP2 as a chromosomal instability gene that regulates Topo IIalpha by sumoylation and suppresses tumorigenesis.


Subject(s)
Antigens, Neoplasm/metabolism , Centromere/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Anaphase , Aneuploidy , Animals , Carcinogens , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Mitosis , Molecular Chaperones/genetics , Mutation , Neoplasms/chemically induced , Neoplasms/metabolism , Nuclear Pore Complex Proteins/genetics , Protein Structure, Tertiary , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
13.
Mol Cell Biol ; 28(10): 3526-37, 2008 May.
Article in English | MEDLINE | ID: mdl-18347063

ABSTRACT

Transcriptional regulation by the canonical Wnt pathway involves the stabilization and nuclear accumulation of beta-catenin, which assembles with LEF1/TCF transcription factors and cofactors to activate Wnt target genes. Recently, the nuclear beta-catenin complex has been shown to contain BCL9, which interacts with beta-catenin and recruits Pygopus as a transcriptional coactivator. However, the presumed general functions of Pygopus and BCL9, which has been proposed to act as a scaffolding protein for Pygopus, have been challenged by the rather specific and modest developmental defects of targeted inactivations of both the Pygo1 and the Pygo2 genes. Here, we analyze the function of BCL9 in transcriptional activation by beta-catenin. We find that BCL9 acts in a cell-type-specific manner and, in part, independent of Pygopus. We show that BCL9 itself contains a transcriptional activation domain in the C terminus, which functionally synergizes in lymphoid cells with the C-terminal transactivation domain of beta-catenin. Finally, we identify amino acids in the transactivation domain of beta-catenin that are important for its function and association with the histone acetyltransferases CBP/p300 and TRRAP/GCN5. Thus, BCL9 may serve to modulate and diversify the transcriptional responses to Wnt signaling in a cell-type-specific manner.


Subject(s)
Neoplasm Proteins/metabolism , Transcriptional Activation , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA Primers/genetics , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Jurkat Cells , Molecular Sequence Data , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA, Small Interfering/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transcription Factors , Transfection , Wnt Proteins/metabolism , beta Catenin/genetics
14.
J Mol Biol ; 374(5): 1200-12, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-17991485

ABSTRACT

The POU domain transcription factor Oct4 plays essential functions in the maintenance of pluripotent embryonic and germ cells of mammals. Molecular mechanisms of Oct4 action remain poorly understood. To isolate modulators of Oct4 activity, we performed a yeast two-hybrid screen with the Oct4 POU domain as a bait and isolated PIASy as an Oct4-interacting protein. Oct4 and PIASy interact in vivo via their POU domain and SAP-domain-containing N terminus, respectively. PIASy does not enhance Oct4 sumoylation but acts as a potent inhibitor of Oct4-mediated transcriptional activation, sequestering Oct4 protein from the vicinity of Cajal bodies and splicing speckles to the nuclear periphery. These modes of PIASy action are uncoupled from its sumoylation activity. Other PIAS family members, PIAS1 and PIAS3, can also interact with Oct4 in vivo and target Oct4 to the nuclear periphery, depending on cellular context. We propose that Oct4 inhibition, mediated by this new class of transcriptional partners, might be instrumental during mammalian development.


Subject(s)
Octamer Transcription Factor-3/physiology , Protein Inhibitors of Activated STAT/physiology , Repressor Proteins/physiology , Animals , Base Sequence , DNA Primers , HeLa Cells , Humans , Immunoprecipitation , Mice , Mutagenesis, Site-Directed , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Protein Binding , Protein Inhibitors of Activated STAT/metabolism , Repressor Proteins/metabolism , Transcriptional Activation
15.
Biochem Biophys Res Commun ; 348(3): 898-907, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16899215

ABSTRACT

The SALL4 promoter has not yet been characterized. Animal studies showed that SALL4 is downstream of and interacts with TBX5 during limb and heart development, but a direct regulation of SALL4 by TBX5 has not been demonstrated. For other SAL genes, regulation within the Shh, Wnt, and Fgf pathways has been reported. Chicken csal1 expression can be activated by a combination of Fgf4 and Wnt3a or Wnt7a. Murine Sall1 enhances, but Xenopus Xsal2 represses, the canonical Wnt signaling. Here we describe the cloning and functional analysis of the SALL4 promoter. Within a minimal promoter region of 31bp, we identified a consensus TCF/LEF-binding site. The SALL4 promoter was strongly activated not only by LEF1 but also by TCF4E. Mutation of the TCF/LEF-binding site resulted in decreased promoter activation. Our results demonstrate for the first time the direct regulation of a SALL gene by the canonical Wnt signaling pathway.


Subject(s)
Gene Expression Regulation/physiology , Promoter Regions, Genetic , Signal Transduction/physiology , TCF Transcription Factors/physiology , Transcription Factors/metabolism , Wnt Proteins/physiology , Animals , Base Sequence , Cell Line , Cell Line, Tumor , Humans , Lymphoid Enhancer-Binding Factor 1/biosynthesis , Lymphoid Enhancer-Binding Factor 1/genetics , Molecular Sequence Data , Transcription Factors/genetics
16.
Mol Cell ; 22(6): 783-794, 2006 06 23.
Article in English | MEDLINE | ID: mdl-16793547

ABSTRACT

Cellular senescence and apoptosis have evolved to restrain unwarranted proliferation of potentially tumorigenic cells. Here we show that overexpression of the E3 SUMO ligase PIASy in normal human fibroblasts recruits the p53 and Rb tumor suppressor pathways to provoke a senescence arrest. By contrast, in Rb-deficient fibroblasts, expression of PIASy leads to p53-dependent apoptosis. Induction of senescence requires PIASy E3 activity and is specific for this member of the PIAS ligase family. PIASy stimulates sumoylation and transcriptional activity of p53 and increases Rb-dependent corepression through recruitment to E2F-responsive promoters. Viral oncoprotein E6 suppresses both PIASy-induced senescence and sumoylation of PIASy substrates. Finally, we show that fibroblasts lacking PIASy exhibit a highly reduced propensity to undergo senescence in response to a prosenescence stimulus. Altogether, these data provide the first evidence for a direct role of an E3 SUMO ligase, and by implication of the SUMO pathway, in cellular senescence and apoptosis.


Subject(s)
Apoptosis/physiology , Cellular Senescence/physiology , Fibroblasts/physiology , Protein Inhibitors of Activated STAT/metabolism , Retinoblastoma Protein/metabolism , Cell Transformation, Viral/genetics , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Fibroblasts/cytology , Gene Expression/genetics , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Poly-ADP-Ribose Binding Proteins , Protein Inhibitors of Activated STAT/deficiency , Protein Processing, Post-Translational , Response Elements/genetics , Retinoblastoma Protein/genetics , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
17.
J Immunol ; 173(10): 6189-99, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15528356

ABSTRACT

Protein inhibitors of activated STATs (PIAS) represent a small family of nuclear proteins that modulate the activity of many transcription factors and act as E3 ligases for covalent modification of proteins with the small ubiquitin-like modifier (SUMO). In particular, PIASy has been shown to inhibit the activation of gene expression by the IFN-responsive transcription factor STAT1 and the Wnt-responsive transcription factor LEF1. To assess the function of PIASy in vivo, we generated and analyzed mice carrying a targeted mutation of the Piasy gene. We find that homozygous mutant mice have no obvious morphological defects and have a normal distribution of lymphocyte populations. Molecular analysis of signaling in response to IFN-gamma and Wnt agonists revealed a modest reduction in the activation of endogenous and transfected target genes. Two-dimensional analysis of total proteins and SUMO-modified proteins in transformed pre-B cells showed no significant differences between wild-type mice and homozygous mutant mice. Taken together, our data indicate that PIASy has a modest effect on cytokine and Wnt signaling, suggesting a redundancy with other members of the family of PIAS proteins.


Subject(s)
Interferons/physiology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Proto-Oncogene Proteins/physiology , Signal Transduction/genetics , Animals , Cell Line, Transformed , Cells, Cultured , Cytokines/physiology , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation, Developmental/genetics , Gene Silencing , Hematopoiesis/genetics , Hematopoiesis/immunology , Homozygote , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins/physiology , Lac Operon , Mice , Mice, Knockout , Mutagenesis, Insertional/methods , Protein Inhibitors of Activated STAT , Signal Transduction/immunology , Wnt Proteins
18.
Genes Dev ; 18(22): 2718-23, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15545629

ABSTRACT

Wnt signaling, which is mediated by LEF1/TCF transcription factors, has been placed upstream of the Notch pathway in vertebrate somitogenesis. Here, we examine the molecular basis for this presumed hierarchy and show that a targeted mutation of Lef1, which abrogates LEF1 function and impairs the activity of coexpressed TCF factors, affects the patterning of somites and the expression of components of the Notch pathway. LEF1 was found to bind multiple sites in the Dll1 promoter in vitro and in vivo. Moreover, mutations of LEF1-binding sites in the Dll1 promoter impair expression of a Dll1-LacZ transgene in the presomitic mesoderm. Finally, the induced expression of LEF1-beta-catenin activates the expression of endogenous Dll1 in fibroblastic cells. Thus, Wnt signaling can affect the Notch pathway by a LEF1-mediated regulation of Dll1.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Membrane Proteins , Membrane Proteins/metabolism , Mutation/genetics , Proto-Oncogene Proteins/metabolism , Somites/physiology , Transcription Factors/physiology , Animals , Body Patterning , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/genetics , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Lac Operon/physiology , Lymphoid Enhancer-Binding Factor 1 , Membrane Proteins/genetics , Mesoderm/cytology , Mesoderm/physiology , Mice , Mice, Knockout , Mice, Transgenic , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Receptors, Notch , Signal Transduction , Somites/cytology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Wnt Proteins , beta Catenin , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...