12 results on '"Kudla AJ"'
Search Results
2. Abstract P6-15-02: Phenotypically distinct HRG positive cancer cells impact standard of care therapies in metastatic breast cancer models
- Author
-
Finn, GJ, primary, Zhang, H, additional, Blois, A, additional, Mathews, SE, additional, Kudla, AJ, additional, Baum, JS, additional, Demars, NA, additional, Cieslewicz, MJ, additional, and Czibere, A, additional
- Published
- 2017
- Full Text
- View/download PDF
3. Single-cell quantitative HER2 measurement identifies heterogeneity and distinct subgroups within traditionally defined HER2-positive patients.
- Author
-
Onsum MD, Geretti E, Paragas V, Kudla AJ, Moulis SP, Luus L, Wickham TJ, McDonagh CF, MacBeath G, and Hendriks BS
- Subjects
- Animals, Breast Neoplasms classification, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, Cluster Analysis, Female, Fluorescent Antibody Technique, Humans, Mice, Mice, Nude, Neoplasms pathology, Reference Standards, Reproducibility of Results, Stomach Neoplasms classification, Stomach Neoplasms metabolism, Stomach Neoplasms pathology, Tissue Array Analysis, Urinary Bladder Neoplasms classification, Urinary Bladder Neoplasms metabolism, Urinary Bladder Neoplasms pathology, Genetic Heterogeneity, Neoplasms classification, Neoplasms metabolism, Receptor, ErbB-2 metabolism, Single-Cell Analysis methods
- Abstract
Human epidermal growth factor receptor 2 (HER2) is an important biomarker for breast and gastric cancer prognosis and patient treatment decisions. HER2 positivity, as defined by IHC or fluorescent in situ hybridization testing, remains an imprecise predictor of patient response to HER2-targeted therapies. Challenges to correct HER2 assessment and patient stratification include intratumoral heterogeneity, lack of quantitative and/or objective assays, and differences between measuring HER2 amplification at the protein versus gene level. We developed a novel immunofluorescence method for quantitation of HER2 protein expression at the single-cell level on FFPE patient samples. Our assay uses automated image analysis to identify and classify tumor versus non-tumor cells, as well as quantitate the HER2 staining for each tumor cell. The HER2 staining level is converted to HER2 protein expression using a standard cell pellet array stained in parallel with the tissue sample. This approach allows assessment of HER2 expression and heterogeneity within a tissue section at the single-cell level. By using this assay, we identified distinct subgroups of HER2 heterogeneity within traditional definitions of HER2 positivity in both breast and gastric cancers. Quantitative assessment of intratumoral HER2 heterogeneity may offer an opportunity to improve the identification of patients likely to respond to HER2-targeted therapies. The broad applicability of the assay was demonstrated by measuring HER2 expression profiles on multiple tumor types, and on normal and diseased heart tissues., (Copyright © 2013 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
4. Antitumor activity of a novel bispecific antibody that targets the ErbB2/ErbB3 oncogenic unit and inhibits heregulin-induced activation of ErbB3.
- Author
-
McDonagh CF, Huhalov A, Harms BD, Adams S, Paragas V, Oyama S, Zhang B, Luus L, Overland R, Nguyen S, Gu J, Kohli N, Wallace M, Feldhaus MJ, Kudla AJ, Schoeberl B, and Nielsen UB
- Subjects
- Animals, Antibodies, Bispecific metabolism, Antibodies, Bispecific pharmacokinetics, Antineoplastic Agents metabolism, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Blotting, Western, Cell Line, Tumor, Cell Proliferation drug effects, Cyclin D1 metabolism, Drug Design, Female, Humans, Inhibitory Concentration 50, Mice, Mice, Inbred NOD, Mice, Nude, Mice, SCID, Multiprotein Complexes metabolism, Neoplasms metabolism, Neoplasms pathology, Phosphatidylinositol 3-Kinases metabolism, Protein Binding, Receptor, ErbB-2 metabolism, Receptor, ErbB-3 metabolism, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Antibodies, Bispecific pharmacology, Neoplasms drug therapy, Neuregulin-1 pharmacology, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-3 antagonists & inhibitors
- Abstract
The prevalence of ErbB2 amplification in breast cancer has resulted in the heavy pursuit of ErbB2 as a therapeutic target. Although both the ErbB2 monoclonal antibody trastuzumab and ErbB1/ErbB2 dual kinase inhibitor lapatinib have met with success in the clinic, many patients fail to benefit. In addition, the majority of patients who initially respond will unfortunately ultimately progress on these therapies. Activation of ErbB3, the preferred dimerization partner of ErbB2, plays a key role in driving ErbB2-amplified tumor growth, but we have found that current ErbB2-directed therapies are poor inhibitors of ligand-induced activation. By simulating ErbB3 inhibition in a computational model of ErbB2/ErbB3 receptor signaling, we predicted that a bispecific antibody that docks onto ErbB2 and subsequently binds to ErbB3 and blocks ligand-induced receptor activation would be highly effective in ErbB2-amplified tumors, with superior activity to a monospecific ErbB3 inhibitor. We have developed a bispecific antibody suitable for both large scale production and systemic therapy by generating a single polypeptide fusion protein of two human scFv antibodies linked to modified human serum albumin. The resulting molecule, MM-111, forms a trimeric complex with ErbB2 and ErbB3, effectively inhibiting ErbB3 signaling and showing antitumor activity in preclinical models that is dependent on ErbB2 overexpression. MM-111 can be rationally combined with trastuzumab or lapatinib for increased antitumor activity and may in the future complement existing ErbB2-directed therapies to treat resistant tumors or deter relapse.
- Published
- 2012
- Full Text
- View/download PDF
5. Therapeutically targeting ErbB3: a key node in ligand-induced activation of the ErbB receptor-PI3K axis.
- Author
-
Schoeberl B, Pace EA, Fitzgerald JB, Harms BD, Xu L, Nie L, Linggi B, Kalra A, Paragas V, Bukhalid R, Grantcharova V, Kohli N, West KA, Leszczyniecka M, Feldhaus MJ, Kudla AJ, and Nielsen UB
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal, Humanized, ErbB Receptors metabolism, Humans, Ligands, Mice, Phosphorylation, Protein Binding, Receptor, ErbB-3 immunology, Signal Transduction, Transplantation, Heterologous, Phosphatidylinositol 3-Kinases metabolism, Receptor, ErbB-3 metabolism
- Abstract
The signaling network downstream of the ErbB family of receptors has been extensively targeted by cancer therapeutics; however, understanding the relative importance of the different components of the ErbB network is nontrivial. To explore the optimal way to therapeutically inhibit combinatorial, ligand-induced activation of the ErbB-phosphatidylinositol 3-kinase (PI3K) axis, we built a computational model of the ErbB signaling network that describes the most effective ErbB ligands, as well as known and previously unidentified ErbB inhibitors. Sensitivity analysis identified ErbB3 as the key node in response to ligands that can bind either ErbB3 or EGFR (epidermal growth factor receptor). We describe MM-121, a human monoclonal antibody that halts the growth of tumor xenografts in mice and, consistent with model-simulated inhibitor data, potently inhibits ErbB3 phosphorylation in a manner distinct from that of other ErbB-targeted therapies. MM-121, a previously unidentified anticancer therapeutic designed using a systems approach, promises to benefit patients with combinatorial, ligand-induced activation of the ErbB signaling network that are not effectively treated by current therapies targeting overexpressed or mutated oncogenes.
- Published
- 2009
- Full Text
- View/download PDF
6. Differential effects of Ras signaling through NFkappaB on skeletal myogenesis.
- Author
-
Mitin N, Kudla AJ, Konieczny SF, and Taparowsky EJ
- Subjects
- Animals, Cell Cycle, Cell Differentiation, Cells, Cultured, DNA-Binding Proteins metabolism, Mice, Myogenic Regulatory Factors metabolism, NF-KappaB Inhibitor alpha, NF-kappa B antagonists & inhibitors, Signal Transduction, Transcription, Genetic, I-kappa B Proteins, Muscle, Skeletal cytology, NF-kappa B metabolism, ras Proteins metabolism
- Abstract
Oncogenic Ras (H-Ras G12V) inhibits skeletal myogenesis through multiple signaling pathways. Previously, we demonstrated that the major downstream effectors of Ras (i.e., MEK/MAPK, RalGDS and Rac/Rho) play a minor, if any, role in the differentiation-defective phenotype of Ras myoblasts. Recently, NFkappaB, another Ras signaling target, has been shown to inhibit myogenesis presumably by stimulating cyclin D1 accumulation and cell cycle progression. In this study, we address the involvement of NFkappaB activation in the Ras-induced inhibition of myogenesis. Using H-Ras G12V and three G12V effector-loop variants, we detect high levels of NFkappaB transcriptional activity in C3H10T1/2-MyoD cells treated with differentiation medium. Myogenesis is blocked by all Ras proteins tested, yet only in the case of H-Ras G12V are cyclin D1 levels increased and cell cycle progression maintained. Expression of IkappaBalpha SR, an inhibitor of NFkappaB, does not reverse the differentiation-defective phenotype of Ras expressing cultures, but does induce differentiation in cultures treated with tumor necrosis factor (TNFalpha) or in cultures expressing the RelA/p65 subunit of NFkappaB. These data confirm that NFkappaB is a target of Ras and suggest that the cellular actions of NFkappaB require additional signals that are discriminated by the Ras effector-loop variants. Results with IkappaBalpha SR convincingly demonstrate that H-Ras G12V does not rely on NFkappaB activity to block myogenesis, an observation that continues to implicate another unidentified signaling pathway(s) in the inhibition of skeletal myogenesis by Ras.
- Published
- 2001
- Full Text
- View/download PDF
7. The FGF receptor-1 tyrosine kinase domain regulates myogenesis but is not sufficient to stimulate proliferation.
- Author
-
Kudla AJ, Jones NC, Rosenthal RS, Arthur K, Clase KL, and Olwin BB
- Subjects
- Animals, Binding Sites, Cell Differentiation physiology, Cell Division, Cell Line, DNA biosynthesis, Humans, Mice, Phosphorylation, Protein-Tyrosine Kinases genetics, Receptor, Fibroblast Growth Factor, Type 1, Receptor, Platelet-Derived Growth Factor beta, Receptors, Fibroblast Growth Factor genetics, Receptors, Platelet-Derived Growth Factor metabolism, Tyrosine metabolism, Muscle, Skeletal cytology, Protein-Tyrosine Kinases metabolism, Receptor Protein-Tyrosine Kinases, Receptors, Fibroblast Growth Factor metabolism
- Abstract
Ligand-stimulated activation of FGF receptors (FGFRs) in skeletal muscle cells represses terminal myogenic differentiation. Skeletal muscle cell lines and subsets of primary cells are dependent on FGFs to repress myogenesis and maintain growth. To understand the intracellular events that transduce these signals, MM14 skeletal muscle cells were transfected with expression vectors encoding chimeric receptors. The chimeras are comprised of the PDGF beta receptor (PDGFbetaR) extracellular domain, the FGFR-1 intracellular domain, and either the PDGFbetaR or FGFR-1 transmembrane domain. The chimeric receptors were autophosphorylated upon PDGF-BB stimulation and are capable of stimulating mitogen-activated protein kinase activity. Activation of the tyrosine kinase domain of either chimera repressed myogenesis, suggesting intracellular responses regulating skeletal muscle differentiation are transduced by activation of the FGFR-1 tyrosine kinase. Unexpectedly, we found that activation of either chimeric receptor failed to stimulate cellular proliferation. Thus, it appears that regulation of skeletal muscle differentiation by FGFs requires only activation of the FGFR tyrosine kinase. In contrast, stimulation of proliferation may require additional, as yet unidentified, signals involving the receptor ectodomain, the FGF ligand, and heparan sulfate either alone, or in combination.
- Published
- 1998
- Full Text
- View/download PDF
8. Muscle LIM protein promotes myogenesis by enhancing the activity of MyoD.
- Author
-
Kong Y, Flick MJ, Kudla AJ, and Konieczny SF
- Subjects
- Animals, Cell Differentiation, Cell Nucleus chemistry, Cytoplasm chemistry, DNA-Binding Proteins metabolism, Dimerization, Helix-Loop-Helix Motifs, LIM Domain Proteins, Mice, Muscle Development, Muscle Proteins analysis, Muscle, Skeletal growth & development, Myogenic Regulatory Factors metabolism, Myogenin metabolism, Protein Binding, Rats, Recombinant Fusion Proteins metabolism, TCF Transcription Factors, Transcription Factor 7-Like 1 Protein, Muscle Proteins metabolism, Muscle, Skeletal cytology, MyoD Protein metabolism, Transcription Factors
- Abstract
The muscle LIM protein (MLP) is a muscle-specific LIM-only factor that exhibits a dual subcellular localization, being present in both the nucleus and in the cytoplasm. Overexpression of MLP in C2C12 myoblasts enhances skeletal myogenesis, whereas inhibition of MLP activity blocks terminal differentiation. Thus, MLP functions as a positive developmental regulator, although the mechanism through which MLP promotes terminal differentiation events remains unknown. While examining the distinct roles associated with the nuclear and cytoplasmic forms of MLP, we found that nuclear MLP functions through a physical interaction with the muscle basic helix-loop-helix (bHLH) transcription factors MyoD, MRF4, and myogenin. This interaction is highly specific since MLP does not associate with nonmuscle bHLH proteins E12 or E47 or with the myocyte enhancer factor-2 (MEF2) protein, which acts cooperatively with the myogenic bHLH proteins to promote myogenesis. The first LIM motif in MLP and the highly conserved bHLH region of MyoD are responsible for mediating the association between these muscle-specific factors. MLP also interacts with MyoD-E47 heterodimers, leading to an increase in the DNA-binding activity associated with this active bHLH complex. Although MLP lacks a functional transcription activation domain, we propose that it serves as a cofactor for the myogenic bHLH proteins by increasing their interaction with specific DNA regulatory elements. Thus, the functional complex of MLP-MyoD-E protein reveals a novel mechanism for both initiating and maintaining the myogenic program and suggests a global strategy for how LIM-only proteins may control a variety of developmental pathways.
- Published
- 1997
- Full Text
- View/download PDF
9. Differentially expressed fibroblast growth factors regulate skeletal muscle development through autocrine and paracrine mechanisms.
- Author
-
Hannon K, Kudla AJ, McAvoy MJ, Clase KL, and Olwin BB
- Subjects
- Actins genetics, Animals, Base Sequence, Cell Differentiation, Cell Division, Cell Line, Fibroblast Growth Factors biosynthesis, Fibroblast Growth Factors classification, Fibroblast Growth Factors genetics, Genes, Reporter, Mice, Molecular Sequence Data, Muscle, Skeletal cytology, Promoter Regions, Genetic, Receptors, Fibroblast Growth Factor physiology, Recombinant Fusion Proteins biosynthesis, Transfection, Fibroblast Growth Factors physiology, Muscle Proteins physiology, Muscle, Skeletal metabolism, Signal Transduction physiology
- Abstract
Several FGF family members are expressed in skeletal muscle; however, the roles of these factors in skeletal muscle development are unclear. We examined the RNA expression, protein levels, and biological activities of the FGF family in the MM14 mouse skeletal muscle cell line. Proliferating skeletal muscle cells express FGF-1, FGF-2, FGF-6, and FGF-7 mRNA. Differentiated myofibers express FGF-5, FGF-7, and reduced levels of FGF-6 mRNA. FGF-3, FGF-4, and FGF-8 were not detectable by RT-PCR in either proliferating or differentiated skeletal muscle cells. FGF-I and FGF-2 proteins were present in proliferating skeletal muscle cells, but undetectable after terminal differentiation. We show that transfection of expression constructs encoding FGF-1 or FGF-2 mimics the effects of exogenously applied FGFs, inhibiting skeletal muscle cell differentiation and stimulating DNA synthesis. These effects require activation of an FGF tyrosine kinase receptor as they are blocked by transfection of a dominant negative mutant FGF receptor. Transient transfection of cells with FGF-1 or FGF-2 expression constructs exerted a global effect on myoblast DNA synthesis, as greater than 50% of the nontransfected cells responded by initiating DNA synthesis. The global effect of cultures transfected with FGF-2 expression vectors was blocked by an anti-FGF-2 monoclonal antibody, suggesting that FGF-2 was exported from the transfected cells. Despite the fact that both FGF-l and FGF-2 lack secretory signal sequences, when expressed intracellularly, they regulate skeletal muscle development. Thus, production of FGF-1 and FGF-2 by skeletal muscle cells may act as a paracrine and autocrine regulator of skeletal muscle development in vivo.
- Published
- 1996
- Full Text
- View/download PDF
10. Conservation of ligand specificity between the mammalian and amphibian fibroblast growth factor receptors.
- Author
-
Patrie KM, Kudla AJ, Olwin BB, and Chiu IM
- Subjects
- 3T3 Cells, Animals, CHO Cells, Cell Differentiation, Cricetinae, DNA, Complementary, Extremities physiology, Mice, Protein Binding, Receptor, Fibroblast Growth Factor, Type 2, Receptors, Fibroblast Growth Factor genetics, Receptors, Fibroblast Growth Factor metabolism, Receptors, Growth Factor genetics, Receptors, Growth Factor metabolism, Receptors, Fibroblast Growth Factor physiology, Receptors, Growth Factor physiology, Regeneration, Salamandridae physiology
- Abstract
We have previously cloned and sequenced a newt keratinocyte growth factor receptor (KGFR) cDNA which exhibited a unique spatial and temporal expression pattern in the regenerating newt limb. In this report, we further characterize the biochemical and functional properties of this newt KGFR. A stable Chinese hamster ovary transfectant overexpressing the newt KGFR was capable of binding both 125I-fibroblast growth factor-1 (FGF-1) and 125I-FGF-7 but not 125I-FGF-2, indistinguishable from the human KGFR. Scatchard analysis and cross-linking studies further support the conclusion that FGF-1 and FGF-7 are the ligands for the newt KGFR. In addition to their ability to bind to FGFs, both the human and the newt KGFR are also capable of repressing differentiation in mouse MM14 myoblasts. MM14 cells express FGFR1 and are repressed from differentiation by FGF-1, FGF-2, and FGF-4 but not FGF-7. Co-transfection of MM14 cells with either a human or newt KGFR expression construct conferred a response to FGF-7 as determined by a human alpha-cardiac actin/luciferase reporter construct. The response to FGF-7 was similar to the endogenous FGF response as FGF-7 prevented MM14 myoblasts from undergoing terminal differentiation. Thus, both the human and the newt KGFRs transduce signals similar to those transduced via the endogenous mouse FGFR1. Together these data indicate that this newly isolated newt KGFR is a functional receptor as it binds two FGF family members with high affinity and mediates signaling in skeletal muscle myoblasts. Because the binding pattern of the newt KGFR is similar to the pattern observed for its mammalian counterpart, it emphasizes the strict conservation that this ligand/receptor system has undergone through evolution.
- Published
- 1995
- Full Text
- View/download PDF
11. A requirement for fibroblast growth factor in regulation of skeletal muscle growth and differentiation cannot be replaced by activation of platelet-derived growth factor signaling pathways.
- Author
-
Kudla AJ, John ML, Bowen-Pope DF, Rainish B, and Olwin BB
- Subjects
- Animals, Cell Differentiation drug effects, Cell Division drug effects, Cell Line, Fibroblast Growth Factors metabolism, Gene Expression Regulation, Mice, Muscle, Skeletal drug effects, Platelet-Derived Growth Factor metabolism, Receptor, Platelet-Derived Growth Factor beta, Second Messenger Systems, Signal Transduction, Fibroblast Growth Factors pharmacology, Muscle Development, Muscle, Skeletal growth & development, Platelet-Derived Growth Factor pharmacology, Receptors, Platelet-Derived Growth Factor metabolism
- Abstract
The distinct effects of cytokines on cellular growth and differentiation suggest that specific signaling pathways mediate these diverse biological activities. Fibroblast growth factors (FGFs) are well-established inhibitors of skeletal muscle differentiation and may operate via activation of specific signaling pathways distinct from recently identified mitogen signaling pathways. We examined whether platelet-derived growth factor (PDGF)-activated signaling pathways are sufficient to mediate FGF-dependent repression of myogenesis by introducing the PDGF beta receptor into a mouse skeletal muscle cell line. Addition of PDGF-BB to cells expressing the PDGF beta receptor activated the PDGF beta receptor tyrosine kinase, stimulated mitogen-activated protein (MAP) kinase, and increased the steady-state levels of junB and c-fos mRNAs. Despite the activation of these intracellular signaling molecules, PDGF beta receptor activation elicited no detectable effect on cell proliferation or differentiation. In contrast to PDGF-BB, addition of FGF-2 to myoblasts activated signaling pathways that resulted in DNA synthesis and repression of differentiation. Because of the low number of endogenous FGF receptors expressed, FGF-stimulated signaling events, including tyrosine phosphorylation and activation of MAP kinase, could be detected only in cells expressing higher levels of a transfected FGF receptor cDNA. As the PDGF beta receptor- and FGF receptor-stimulated signaling pathways yield different biological responses in these skeletal muscle cells, we hypothesize that FGF-mediated repression of skeletal muscle differentiation activates signaling pathways distinct from those activated by the PDGF beta receptor. Activation of PDGF beta receptor tyrosine kinase activity, stimulation of MAP kinase, and upregulation of immediate-early gene expression are not sufficient to repress skeletal muscle differentiation.
- Published
- 1995
- Full Text
- View/download PDF
12. Are fibroblast growth factors regulators of myogenesis in vivo?
- Author
-
Olwin BB, Hannon K, and Kudla AJ
- Subjects
- Animals, Cell Differentiation physiology, Fibroblast Growth Factors analogs & derivatives, Fibroblast Growth Factors analysis, Humans, Muscle, Skeletal chemistry, Receptors, Fibroblast Growth Factor analysis, Receptors, Fibroblast Growth Factor physiology, Fibroblast Growth Factors physiology, Muscle, Skeletal embryology
- Abstract
Recent advances in understanding of skeletal muscle differentiation implicate fibroblast growth factors (FGFs) as regulators of myogenesis; however, the identity and actions of factors that repress myogenesis in vivo remain to be established. This review will focus on the fibroblast growth factor family and the evidence for its role in regulating myogenesis in culture and in vivo.
- Published
- 1994
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.