Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
BMC Biotechnol ; 15: 95, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26475607

ABSTRACT

BACKGROUND: Amidation of the carboxyl terminal of many peptides is essential for full biological potency, often increasing receptor binding and stability. The single enzyme responsible for this reaction is peptidylglycine α-amidating monooxygenase (PAM: EC 1.14.17.3), a copper- and ascorbate-dependent Type I membrane protein. METHODS: To make large amounts of high molecular weight amidated product, Chinese hamster ovary (CHO) cells were engineered to express exogenous PAM. To vary access of the enzyme to its substrate, exogenous PAM was targeted to the endoplasmic reticulum, trans-Golgi network, endosomes and lysosomes or to the lumen of the secretory pathway. RESULTS: PAM was equally active when targeted to each intracellular location and assayed in homogenates. Immunocytochemical analyses of CHO cells and a pituitary cell line demonstrated that targeting of exogenous PAM was partially successful. PAM substrates generated by expressing peptidylglycine substrates (glucagon-like peptide 1-Gly, peptide YY-Gly and neuromedin U-Gly) fused to the C-terminus of immunoglobulin Fc in CHO cell lines producing targeted PAM. The extent of amidation of the Fc-peptides was determined by mass spectrometry and amidation-specific enzyme immunoassays. Amidation was inhibited by copper chelation, but was not enhanced by the addition of additional copper or ascorbate. CONCLUSIONS: Peptide amidation was increased over endogenous levels by exogenous PAM, and targeting PAM to the endoplasmic reticulum or trans-Golgi network increased peptide amidation compared to endogenous CHO PAM.


Subject(s)
Immunoglobulin Fc Fragments/metabolism , Metabolic Engineering/methods , Peptides/metabolism , Recombinant Fusion Proteins/metabolism , Amides/chemistry , Amides/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/analysis , Immunoglobulin Fc Fragments/chemistry , Mass Spectrometry , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Peptides/analysis , Peptides/chemistry , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/chemistry
2.
BMC Biotechnol ; 15: 61, 2015 Jun 27.
Article in English | MEDLINE | ID: mdl-26116580

ABSTRACT

BACKGROUND: The therapeutic use of α-amidated peptides (e.g. calcitonin, glucagon-like peptide) has increased dramatically, but there are major impediments to wider use of such peptides. Larger peptides are expensive to synthesize, and short plasma half-lives frequently limit the clinical circumstances in which the peptides would be useful. Both problems are potentially solved by producing peptides as fusions with the Fc region of human immunoglobulin. METHODS: Glucagon-like peptide 1 (GLP1), peptide YY (PYY) and neuromedin U (NMU) were expressed and purified from stable CHO lines; since the α-amide group is essential for full biological potency of many peptides, Fc-fusion peptides were expressed in CHO lines stably expressing the α-amidating enzyme, peptidylglycine α-amidating monooxygenase (PAM: EC 1.14.17.3). Purified fusion proteins were analyzed intact and after HRV3C rhinovirus protease cleavage, at a site in the linker separating the Fc region from the peptide, by mass spectrometry and amide-specific immunoassays. RESULTS: The Fc fusions were expressed at 1-2.5 µg/mg cell protein and secreted at 5-20% of cell content per hour, in a peptide-specific manner. CHO cells express measurable endogenous PAM activity, amidating 25% of Fc-PYY and almost 90% of Fc-GLP1. Expression of exogenous PAM increased the level of peptide amidation to 50% of Fc-PYY and 95 % of Fc-NMU. The Fc-GLP1 fusions were 10,000-fold less active than synthetic GLP1 in a cell-receptor cyclic AMP-based assay, as expected since the amino terminal of GLP1 is essential for full biological activity. The Fc-PYY fusions were 100-fold less active than PYY-NH2 but 10-fold more active than non-amidated PYY-Gly. CONCLUSIONS: This type of approach can be used for the production of stabilized α-amidated peptides aimed at clinical trials.


Subject(s)
Amides/metabolism , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Amides/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Glucagon-Like Peptide 1 , Humans , Immunoglobulin Fc Fragments/genetics , Neuropeptides , Peptide YY , Protein Stability , Recombinant Fusion Proteins/genetics
3.
Methods ; 65(1): 114-26, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23872058

ABSTRACT

The Fc variant of IgG2, designated as IgG2σ, was engineered with V234A/G237A /P238S/H268A/V309L/A330S/P331S substitutions to eliminate affinity for Fcγ receptors and C1q complement protein and consequently, immune effector functions. IgG2σ was compared to other previously well-characterized Fc 'muted' variants, including aglycosylated IgG1, IgG2m4 (H268Q/V309L/A330S/P331S, changes to IgG4), and IgG4 ProAlaAla (S228P/L234A/L235A) in its capacity to bind FcγRs and activate various immune-stimulatory responses. In contrast to the previously characterized muted Fc variants, which retain selective FcγR binding and effector functions, IgG2σ shows no detectable binding to the Fcγ receptors in affinity and avidity measurements, nor any detectable antibody-dependent cytotoxicity, phagocytosis, complement activity, or Fc-mediated cytokine release. Moreover, IgG2σ shows minimal immunogenic potential by T-cell epitope analysis. The circulating half-life of IgG2σ in monkeys is extended relative to IgG1 and IgG2, in spite of similar in vitro binding to recombinant FcRn. The three-dimensional structure of the Fc, needed for assessing the basis for the absence of effector function, was compared with that of IgG2 revealing a number of conformational differences near the hinge region of the CH2 domain that result from the amino acid substitutions. Modeling reveals that at least one of the key interactions with FcγRs is disrupted by a conformational change that reorients P329 to a position that prevents it from interacting with conserved W90 and W113 residues of the FcγRs. Inspection of the structure also indicated significant changes to the conformations of D270 and P329 in the CH2 domain that could negatively impact C1q binding. Thus, structural perturbations of the Fc provide a rationale for the loss of function. In toto, these properties of IgG2σ suggest that it is a superior alternative to previously described IgG variants of minimal effector function, for future therapeutic applications of non-immunostimulatory mAb and Fc-fusion platforms.


Subject(s)
Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Immunologic Factors/chemistry , Amino Acid Substitution , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , Binding Sites , Crystallography, X-Ray , Cytokines/metabolism , HEK293 Cells , Half-Life , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/genetics , Immunoglobulin G/pharmacology , Immunologic Factors/genetics , Immunologic Factors/pharmacology , Macaca fascicularis , Male , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Secondary , Receptor, ErbB-2/immunology , Receptors, IgG/chemistry
4.
J Mol Recognit ; 25(3): 147-54, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22407978

ABSTRACT

Immunoglobulin G (IgG) antibodies are an integral part of the adaptive immune response that provide a direct link between humoral and cellular components of the immune system. Insights into relationships between the structure and function of human IgGs have prompted molecular engineering efforts to enhance or eliminate specific properties, such as Fc-mediated immune effector functions. Human IgGs have an N-glycosylation site at Asn297, located in the second heavy chain constant region (CH2). The composition of the Fc glycan can have substantial impacts on Fc gamma receptor(FcγR) binding. The removal of the glycan through enzymatic deglycosylation or mutagenesis of the N-linked glycosylation site has been reported to "silence" FcγR-binding and effector functions, particularly with assays that measure monomeric binding. However, interactions between IgGs and FcγRs are not limited to monomeric interactions but can be influenced by avidity, which takes into account the sum of multimeric interactions between antigen-engaged IgGs and FcγRs. We show here that under in vitro conditions, which allowed avidity binding, aglycosylated IgGs can bind to one of the FcγRs, FcγRI, and mediate effector functions. These studies highlight how the valency of a molecular interaction (monomeric binding versus avidity binding) can influence antibody/FcγR interactions such that avidity effects can translate very low intrinsic affinities into significant functional outcomes.


Subject(s)
Antibody Affinity , Immunoglobulin G/metabolism , Receptors, IgG/metabolism , Animals , Antigens, CD/immunology , Binding, Competitive , Cell Proliferation , Glycosylation , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Macrophages/immunology , Macrophages/physiology , Mice , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/chemistry , Phagocytosis/immunology , Protein Binding , Receptors, IgG/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/physiology
5.
Cancer Cell ; 1(5): 493-503, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12124178

ABSTRACT

Chemotherapeutic agents simultaneously induce transcription factors p53 and NFkappaB. p53 induction can activate an apoptotic program, and resistance to chemotherapy correlates with the loss of a functional p53 pathway. By contrast, NFkappaB prevents apoptosis in response to chemotherapeutic agents. We have analyzed the p53 response in IKK1/2(-/-) MEFs, which lack detectable NFkappaB activity. Compared to WT fibroblasts, IKK1/2(-/-) fibroblasts showed increased cell death and p53 induction in response to the chemotherapeutic agent, doxorubicin. Reconstitution of IKK2, but not IKK1, increased Mdm2 levels and decreased doxorubicin-induced p53 stabilization and cell death. IKK2-mediated effects required its kinase function and were abrogated by coexpression of the dominant negative IkappaBalphaM, implying a role for NFkappaB in blocking chemotherapy-induced p53 and cell death.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/physiology , Doxorubicin/pharmacology , NF-kappa B/metabolism , Nuclear Proteins , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Down-Regulation , Drug Resistance, Neoplasm , Enzyme Stability , Fibroblasts/cytology , I-kappa B Kinase , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-mdm2 , RNA, Messenger/metabolism , Retroviridae , Tumor Suppressor Protein p53/genetics , Up-Regulation
6.
Proc Natl Acad Sci U S A ; 106(42): 17864-9, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19815504

ABSTRACT

The successful elimination of pathogenic cells and microorganisms by the humoral immune system relies on effective interactions between host immunoglobulins and Fc gamma receptors on effector cells, in addition to the complement system. Essential Ig motifs that direct those interactions reside within the conserved IgG lower hinge/CH2 interface. We noted that a group of tumor-related and microbial proteases cleaved human IgG1s in that region, and the "nick" of just one of the heavy chains profoundly inhibited IgG1 effector functions. We focused on IgG1 monoclonal antibodies (mAbs) since IgG1 is the most abundant human subclass and demonstrates robust Fc-mediated effector functions. The loss of Fc-mediated cell killing activities was correlated with diminished binding to the Fc gamma family of receptors, but a similar decrease in affinity was not observed toward the FcRn receptor that maintains IgG in circulation. Endogenous human IgG cleavage products of comparable size to mAbs with the single cleavage were detected by Western blot analysis in synovial fluid from patients with rheumatoid arthritis and in breast carcinoma extracts. Their detection is problematic under physiological conditions, since there is no loss of structure, and antigen-binding capability is unaffected. These findings suggest that within the hostile proteolytic microenvironments associated with many diseases, key effector functions of host IgGs, or therapeutic Abs, may be compromised.


Subject(s)
Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Peptide Hydrolases/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , Bacterial Proteins/metabolism , Binding Sites , Breast Neoplasms/enzymology , Cell Membrane/immunology , Female , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/metabolism , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Receptors, IgG/metabolism , Serine Endopeptidases/metabolism , Staphylococcus aureus/enzymology , Streptococcus pyogenes/enzymology
7.
Biologicals ; 39(1): 9-22, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20888784

ABSTRACT

To assess the impact of manufacturing changes on antibody structure and function during the course of product development, three comparability studies were performed for each of two different IgG1 monoclonal antibody product candidates. Comparability study #1 evaluated the effect of changing the cell line and bulk drug substance manufacturing process for cell culture and purification. Results indicated that these process changes led to differences in sialylation of N-glycans and/or C-terminal lysine levels. Comparability study #2 results confirmed that scale-up of the bulk process and transfer to the commercial site, combined with changing from a lyophilized to a liquid dosage form, did not impact the structural or functional integrity of the antibodies. Comparability study #3 examined possible differences arising when the liquid formulation filled into pre-filled syringes and vials. Results indicated nearly identical molecular structure, biological activity, and degradation profiles except for a small yet statistically significant increase in the levels of subvisible particles in pre-filled syringes. These results from comparability studies with two different monoclonal antibodies are discussed with respect to the timing of the manufacturing changes and overall comparability strategies to assure safety and efficacy during development.


Subject(s)
Antibodies, Monoclonal/analysis , Drug Industry/standards , Immunoglobulin G/immunology , Technology, Pharmaceutical/standards , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Cell Line , Chromatography, High Pressure Liquid , Circular Dichroism , Drug Industry/methods , Electrophoresis, Polyacrylamide Gel , Humans , K562 Cells , Protein Binding , Receptors, IgG/metabolism , Technology, Pharmaceutical/methods
8.
Front Oncol ; 10: 446, 2020.
Article in English | MEDLINE | ID: mdl-32351885

ABSTRACT

This perspective highlights the history and challenges of developing CD3-based bispecific T-cell engagers (TCEs) as cancer therapeutics as well as considerations and potential strategies for designing the next generation TCE molecules. The goal of this article is to raise awareness of natural T-cell biology and how to best harness the tumor cell killing capacity of cytotoxic T-cells with TCEs. In light of 30 years of concerted efforts to advance TCEs in early clinical development, many of the first-generation bispecific antibodies have exhibited lackluster safety, efficacy, and manufacturability profiles. As of January 2020, blinatumomab remains the only approved TCE. Many of the current set-backs in early clinical trials implicate the high-affinity CD3 binding domains employed and the respective bispecific platforms as potential culprits. The underlying conviction of the authors is that by taking corrective measures, TCEs can transform cancer therapy. Through openness, transparency, and much needed feedback from ongoing clinical studies, the field can continuously improve the design and effectiveness of next generation T-cell redirecting therapeutics.

9.
MAbs ; 11(4): 639-652, 2019.
Article in English | MEDLINE | ID: mdl-30698484

ABSTRACT

T-cell-recruiting bispecific antibodies (T-BsAbs) have shown potent tumor killing activity in humans, but cytokine release-related toxicities have affected their clinical utility. The use of novel anti-CD3 binding domains with more favorable properties could aid in the creation of T-BsAbs with improved therapeutic windows. Using a sequence-based discovery platform, we identified new anti-CD3 antibodies from humanized rats that bind to multiple epitopes and elicit varying levels of T-cell activation. In T-BsAb format, 12 different anti-CD3 arms induce equivalent levels of tumor cell lysis by primary T-cells, but potency varies by a thousand-fold. Our lead CD3-targeting arm stimulates very low levels of cytokine release, but drives robust tumor antigen-specific killing in vitro and in a mouse xenograft model. This new CD3-targeting antibody underpins a next-generation T-BsAb platform in which potent cytotoxicity is uncoupled from high levels of cytokine release, which may lead to a wider therapeutic window in the clinic.


Subject(s)
Antibodies, Bispecific/metabolism , Antibodies, Monoclonal/metabolism , CD3 Complex/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Animals, Inbred Strains , Antigens, Neoplasm/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic , Female , Humans , Jurkat Cells , Lymphocyte Activation , Mice , Neoplasms/immunology , Rats , Xenograft Model Antitumor Assays
10.
Front Immunol ; 9: 889, 2018.
Article in English | MEDLINE | ID: mdl-29740455

ABSTRACT

We created a novel transgenic rat that expresses human antibodies comprising a diverse repertoire of heavy chains with a single common rearranged kappa light chain (IgKV3-15-JK1). This fixed light chain animal, called OmniFlic, presents a unique system for human therapeutic antibody discovery and a model to study heavy chain repertoire diversity in the context of a constant light chain. The purpose of this study was to analyze heavy chain variable gene usage, clonotype diversity, and to describe the sequence characteristics of antigen-specific monoclonal antibodies (mAbs) isolated from immunized OmniFlic animals. Using next-generation sequencing antibody repertoire analysis, we measured heavy chain variable gene usage and the diversity of clonotypes present in the lymph node germinal centers of 75 OmniFlic rats immunized with 9 different protein antigens. Furthermore, we expressed 2,560 unique heavy chain sequences sampled from a diverse set of clonotypes as fixed light chain antibody proteins and measured their binding to antigen by ELISA. Finally, we measured patterns and overall levels of somatic hypermutation in the full B-cell repertoire and in the 2,560 mAbs tested for binding. The results demonstrate that OmniFlic animals produce an abundance of antigen-specific antibodies with heavy chain clonotype diversity that is similar to what has been described with unrestricted light chain use in mammals. In addition, we show that sequence-based discovery is a highly effective and efficient way to identify a large number of diverse monoclonal antibodies to a protein target of interest.


Subject(s)
Antibodies, Monoclonal/immunology , Drug Discovery/methods , Genes, Immunoglobulin Heavy Chain/genetics , Genes, Immunoglobulin Light Chain/genetics , Immunoglobulin kappa-Chains/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antigens/administration & dosage , Antigens/immunology , B-Lymphocytes/immunology , Germinal Center/cytology , Germinal Center/immunology , High-Throughput Nucleotide Sequencing , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin kappa-Chains/genetics , Models, Animal , Rats , Rats, Sprague-Dawley , Rats, Transgenic
11.
Methods Mol Biol ; 841: 321-49, 2012.
Article in English | MEDLINE | ID: mdl-22222459

ABSTRACT

Antibodies make up the largest, growing segment of protein therapeutics in the pharmaceutical and biotechnology industries. The development or engineering of therapeutic antibodies is based to a large extent on our knowledge of antibody structure and requires sophisticated methods that continue to evolve. In this chapter, after a review of what is known about the structure and functional properties of antibodies, the current, state-of-the-art antibody engineering methods are described. These methods include antibody humanization, antigen-affinity optimization, Fc engineering for modulated effector function and extended half-life, and engineering for improved stability and biophysical properties. X-ray crystallographic structures of antibody fragments and their complexes can play a critical role in guiding and, in some cases, accelerating these processes. These approaches represent guidelines for developing antibody therapeutics with the desired affinity, effector function, and biophysical properties.


Subject(s)
Antibodies/therapeutic use , Drug Design , Protein Engineering , Antibodies/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure
12.
MAbs ; 2(4): 428-39, 2010.
Article in English | MEDLINE | ID: mdl-20519961

ABSTRACT

We prepared and characterized golimumab (CNTO148), a human IgG1 tumor necrosis factor alpha (TNFα) antagonist monoclonal antibody chosen for clinical development based on its molecular properties. Golimumab was compared with infliximab, adalimumab and etanercept for affinity and in vitro TNFα neutralization. The affinity of golimumab for soluble human TNFα, as determined by surface plasmon resonance, was similar to that of etanercept (18 pM versus 11 pM), greater than that of infliximab (44 pM) and significantly greater than that of adalimumab (127 pM, p=0.018).  The concentration of golimumab necessary to neutralize TNFα-induced E-selectin expression on human endothelial cells by 50% was significantly less than those for infliximab (3.2 fold; p=0.017) and adalimumab (3.3-fold; p=0.008) and comparable to that for etanercept. The conformational stability of golimumab was greater than that of infliximab (primary melting temperature [Tm] 74.8 °C vs. 69.5 °C) as assessed by differential scanning calorimetry.  In addition, golimumab showed minimal aggregation over the intended shelf life when formulated as a high concentration liquid product (100 mg/mL) for subcutaneous administration.  In vivo, golimumab at doses of 1 and 10 mg/kg significantly delayed disease progression in a mouse model of human TNFα-induced arthritis when compared with untreated mice, while infliximab was effective only at 10 mg/kg. Golimumab also significantly reduced histological scores for arthritis severity and cartilage damage, as well as serum levels of pro-inflammatory cytokines and chemokines associated with arthritis. Thus, we have demonstrated that golimumab is a highly stable human monoclonal antibody with high affinity and capacity to neutralize human TNFα in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/pharmacology , Arthritis/immunology , Cartilage/drug effects , Immunoglobulin G/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Adalimumab , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Affinity , Arthritis/chemically induced , Cartilage/pathology , Disease Models, Animal , Disease Progression , E-Selectin/genetics , E-Selectin/metabolism , Etanercept , Gene Expression Regulation/drug effects , Hybridomas , Immunoglobulin G/isolation & purification , Inflammation Mediators/metabolism , Infliximab , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Protein Conformation , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/immunology
13.
Proc Natl Acad Sci U S A ; 103(11): 4005-10, 2006 Mar 14.
Article in English | MEDLINE | ID: mdl-16537476

ABSTRACT

Antibody-dependent cell-mediated cytotoxicity, a key effector function for the clinical efficacy of monoclonal antibodies, is mediated primarily through a set of closely related Fcgamma receptors with both activating and inhibitory activities. By using computational design algorithms and high-throughput screening, we have engineered a series of Fc variants with optimized Fcgamma receptor affinity and specificity. The designed variants display >2 orders of magnitude enhancement of in vitro effector function, enable efficacy against cells expressing low levels of target antigen, and result in increased cytotoxicity in an in vivo preclinical model. Our engineered Fc regions offer a means for improving the next generation of therapeutic antibodies and have the potential to broaden the diversity of antigens that can be targeted for antibody-based tumor therapy.


Subject(s)
Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Alemtuzumab , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/metabolism , Antibody Affinity , Antibody Specificity , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/metabolism , B-Lymphocytes/immunology , Complement System Proteins/metabolism , Cytotoxicity, Immunologic , Genetic Variation , Humans , In Vitro Techniques , Lymphocyte Depletion , Macaca fascicularis , Protein Engineering , Receptors, IgG/metabolism , Trastuzumab
14.
Mol Cell ; 9(5): 1031-44, 2002 May.
Article in English | MEDLINE | ID: mdl-12049739

ABSTRACT

Oncogene overexpression activates p53 by a mechanism posited to involve uncharacterized hyperproliferative signals. We determined whether such signals produce metabolic perturbations that generate DNA damage, a known p53 inducer. Biochemical, cytological, cell cycle, and global gene expression analyses revealed that brief c-Myc activation can induce DNA damage prior to S phase in normal human fibroblasts. Damage correlated with induction of reactive oxygen species (ROS) without induction of apoptosis. Deregulated c-Myc partially disabled the p53-mediated DNA damage response, enabling cells with damaged genomes to enter the cycle, resulting in poor clonogenic survival. An antioxidant reduced ROS, decreased DNA damage and p53 activation, and improved survival. We propose that oncogene activation can induce DNA damage and override damage controls, thereby accelerating tumor progression via genetic instability.


Subject(s)
DNA Damage , Proto-Oncogene Proteins c-myc/pharmacology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Cells, Cultured , Fibroblasts/metabolism , Gene Expression Regulation , Humans , In Situ Nick-End Labeling
15.
Science ; 301(5641): 1895-8, 2003 Sep 26.
Article in English | MEDLINE | ID: mdl-14512626

ABSTRACT

Tumor necrosis factor (TNF) is a key regulator of inflammatory responses and has been implicated in many pathological conditions. We used structure-based design to engineer variant TNF proteins that rapidly form heterotrimers with native TNF to give complexes that neither bind to nor stimulate signaling through TNF receptors. Thus, TNF is inactivated by sequestration. Dominant-negative TNFs represent a possible approach to anti-inflammatory biotherapeutics, and experiments in animal models show that the strategy can attenuate TNF-mediated pathology. Similar rational design could be used to engineer inhibitors of additional TNF superfamily cytokines as well as other multimeric ligands.


Subject(s)
Protein Engineering , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Substitution , Animals , Antigens, CD/metabolism , Apoptosis , Arthritis, Experimental/drug therapy , Biopolymers , Caspases/metabolism , Cell Line , Cell Nucleus/metabolism , Computer Simulation , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Galactosamine/pharmacology , HeLa Cells , Humans , Liver/drug effects , NF-kappa B/metabolism , Point Mutation , Rats , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Transcription Factor RelA , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL