Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 387(2): 214-225, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37643795

ABSTRACT

Hereditary angioedema (HAE) is a rare autosomal dominant disorder caused by a deficiency in functional C1 esterase inhibitor, a serpin family protein that blocks the activity of plasma kallikrein. Insufficient inhibition of plasma kallikrein results in the overproduction of bradykinin, a vasoactive inflammatory mediator that produces both pain and unpredictable swelling during HAE attacks, with potentially life-threatening consequences. We describe the generation of STAR-0215, a humanized IgG1 antibody with a long circulating half-life (t1/2) that potently inhibits plasma kallikrein activity, with a >1000-fold lower affinity for prekallikrein and no measurable inhibitory activity against other serine proteases. The high specificity and inhibitory effect of STAR-0215 is demonstrated through a unique allosteric mechanism involving N-terminal catalytic domain binding, destabilization of the activation domain, and reversion of the active site to the inactive zymogen state. The YTE (M252Y/S254T/T256E) modified fragment crystallizable (Fc) domain of STAR-0215 enhances pH-dependent neonatal Fc receptor binding, resulting in a prolonged t1/2 in vivo (∼34 days in cynomolgus monkeys) compared with antibodies without this modification. A single subcutaneous dose of STAR-0215 (≥100 mg) was predicted to be active in patients for 3 months or longer, based on simulations using a minimal physiologically based pharmacokinetic model. These data indicate that STAR-0215, a highly potent and specific antibody against plasma kallikrein with extended t1/2, is a potential agent for long-term preventative HAE therapy administered every 3 months or less frequently. SIGNIFICANCE STATEMENT: STAR-0215 is a YTE-modified immunoglobulin G1 monoclonal antibody with a novel binding mechanism that specifically and potently inhibits the enzymatic activity of plasma kallikrein and prevents the generation of bradykinin. It has been designed to be a long-lasting prophylactic treatment to prevent attacks of HAE and to decrease the burden of disease and the burden of treatment for people with HAE.

2.
J Med Chem ; 59(23): 10781-10787, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27809515

ABSTRACT

Herein we report structure-cytotoxicity relationships for analogues of N14-desacetoxytubulyisn H 1. A novel synthetic approach toward 1 enabled the discovery of compounds with a range of activity. Calculated basicity of the N-terminus of tubulysins was shown to be a good predictor of cytotoxicity. The impact of structural modifications at the C-terminus of 1 upon cytotoxicity is also described. These findings will facilitate the development of new tubulysin analogues for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Oligopeptides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Structure-Activity Relationship
3.
Oncotarget ; 7(42): 68278-68291, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27626702

ABSTRACT

Antibodies that target cell-surface molecules on T cells can enhance anti-tumor immune responses, resulting in sustained immune-mediated control of cancer. We set out to find new cancer immunotherapy targets by phenotypic screening on human regulatory T (Treg) cells and report the discovery of novel activators of tumor necrosis factor receptor 2 (TNFR2) and a potential role for this target in immunotherapy. A diverse phage display library was screened to find antibody mimetics with preferential binding to Treg cells, the most Treg-selective of which were all, without exception, found to bind specifically to TNFR2. A subset of these TNFR2 binders were found to agonise the receptor, inducing iκ-B degradation and NF-κB pathway signalling in vitro. TNFR2 was found to be expressed by tumor-infiltrating Treg cells, and to a lesser extent Teff cells, from three lung cancer patients, and a similar pattern was also observed in mice implanted with CT26 syngeneic tumors. In such animals, TNFR2-specific agonists inhibited tumor growth, enhanced tumor infiltration by CD8+ T cells and increased CD8+ T cell IFN-γ synthesis. Together, these data indicate a novel mechanism for TNF-α-independent TNFR2 agonism in cancer immunotherapy, and demonstrate the utility of target-agnostic screening in highlighting important targets during drug discovery.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Receptors, Tumor Necrosis Factor, Type II/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Female , HEK293 Cells , Humans , Jurkat Cells , Mice, Inbred BALB C , NF-kappa B/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/therapy , Phenotype , Receptors, Tumor Necrosis Factor, Type II/agonists , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects
4.
Cancer Immunol Res ; 3(9): 1052-62, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25943534

ABSTRACT

Programmed cell-death 1 ligand 1 (PD-L1) is a member of the B7/CD28 family of proteins that control T-cell activation. Many tumors can upregulate expression of PD-L1, inhibiting antitumor T-cell responses and avoiding immune surveillance and elimination. We have identified and characterized MEDI4736, a human IgG1 monoclonal antibody that binds with high affinity and specificity to PD-L1 and is uniquely engineered to prevent antibody-dependent cell-mediated cytotoxicity. In vitro assays demonstrate that MEDI4736 is a potent antagonist of PD-L1 function, blocking interaction with PD-1 and CD80 to overcome inhibition of primary human T-cell activation. In vivo MEDI4736 significantly inhibits the growth of human tumors in a novel xenograft model containing coimplanted human T cells. This activity is entirely dependent on the presence of transplanted T cells, supporting the immunological mechanism of action for MEDI4736. To further determine the utility of PD-L1 blockade, an anti-mouse PD-L1 antibody was investigated in immunocompetent mice. Here, anti-mouse PD-L1 significantly improved survival of mice implanted with CT26 colorectal cancer cells. The antitumor activity of anti-PD-L1 was enhanced by combination with oxaliplatin, which resulted in increased release of HMGB1 within CT26 tumors. Taken together, our results demonstrate that inhibition of PD-L1 function can have potent antitumor activity when used as monotherapy or in combination in preclinical models, and suggest it may be a promising therapeutic approach for the treatment of cancer. MEDI4736 is currently in several clinical trials both alone and in combination with other agents, including anti-CTLA-4, anti-PD-1, and inhibitors of IDO, MEK, BRAF, and EGFR.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-1 Antigen/metabolism , B7-H1 Antigen/metabolism , Binding, Competitive , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Melanoma/immunology , Melanoma/pathology , Melanoma/prevention & control , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/prevention & control , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Mol Cancer Ther ; 13(2): 386-98, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24344235

ABSTRACT

The hedgehog pathway has been implicated in the tumorigenesis, tumor progression, and metastasis of numerous human cancers. We generated the first fully human hedgehog antibody MEDI-5304 and characterized its antitumor activity and preclinical toxicology. MEDI-5304 bound sonic hedgehog (SHH) and Indian hedgehog (IHH) with low picomolar affinity and neutralized SHH and IHH activity in cellular mGLI1 reporter assays. The antibody inhibited transcription of hedgehog target genes and osteoblast differentiation of C3H10T1/2 cells. We evaluated the activity of MEDI-5304 in vivo in model systems that allowed us to evaluate two primary hypotheses of hedgehog function in human cancer, paracrine signaling between tumor and stromal cells and cancer stem cell (CSC) self-renewal. MEDI-5304 displayed robust pharmacodynamic effects in stromal cells that translated to antitumor efficacy as a single agent in an HT-29/MEF coimplantation model of paracrine hedgehog signaling. MEDI-5304 also improved responses to carboplatin in the HT-29/MEF model. The antibody, however, had no effect as a single agent or in combination with gemcitabine on the CSC frequency or growth of several primary pancreatic cancer explant models. These findings support the conclusion that hedgehog contributes to tumor biology via paracrine tumor-stromal signaling but not via CSC maintenance or propagation. Finally, the only safety study finding associated with MEDI-5304 was ondontodysplasia in rats. Thus, MEDI-5304 represents a potent dual hedgehog inhibitor suitable for continued development to evaluate efficacy and safety in human patients with tumors harboring elevated levels of SHH or IHH.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Paracrine Communication/drug effects , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Female , HT29 Cells , Hedgehog Proteins/immunology , Humans , Kinetics , Macaca fascicularis , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , NIH 3T3 Cells , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Paracrine Communication/immunology , Protein Binding/immunology , Rats, Wistar , Stromal Cells/drug effects , Stromal Cells/immunology , Stromal Cells/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Mol Pharmacol ; 83(6): 1247-56, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23558446

ABSTRACT

Platelet-derived growth factor receptor α (PDGFRα) is a receptor tyrosine kinase that promotes cell survival and is expressed in both the tumor and the stromal components of human cancers. We have developed a fully human monoclonal antibody, MEDI-575, that selectively binds to human PDGFRα with high affinity, with no observable affinity for murine PDGFRα. To more fully characterize the role of PDGFRα in the regulation of tumor stroma, we evaluated the in vivo antitumor effects of MEDI-575 in tumor-bearing severe combined immunodeficient (SCID) mice and in genetically altered SCID mice expressing human PDGFRα in place of murine PDGFRα. We used the Calu-6 non-small cell lung cancer model because it lacks an in vitro proliferative response to PDGFRα activation. Antitumor activity was observed when the study was performed in mice expressing the human receptor, but no activity was observed in the mice expressing the murine receptor. Immunohistologic analysis of the tumors from mice expressing human PDGFRα showed a highly significant reduction in stromal fibroblast content and only minor changes in tumor proliferative index in tumors exposed to MEDI-575 compared with the results seen in vehicle-treated tumors or in tumors from mice expressing murine PDGFRα. Additional in vitro studies indicated that exposure of primary cancer-associated fibroblasts to MEDI-575 can directly affect proliferation and key signaling pathways in these cells. These results highlight the potential for observing antitumor activity with MEDI-575 through modulation of the stromal component of tumors and confirm that the PDGFRα pathway can play a role in maintaining a tumor microenvironment conducive to tumor growth.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Fibroblasts/drug effects , Lung Neoplasms/pathology , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Nude , Mice, SCID , NIH 3T3 Cells , Neoplasm Transplantation , Phosphorylation , Receptor, Platelet-Derived Growth Factor alpha/genetics , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Transplantation, Heterologous
7.
MAbs ; 5(2): 255-62, 2013.
Article in English | MEDLINE | ID: mdl-23412563

ABSTRACT

Antibodies can undergo a variety of covalent and non-covalent degradation reactions that have adverse effects on efficacy, safety, manufacture and storage. We had identified an antibody to Angiopoietin 2 (Ang2 mAb) that neutralizes Ang2 binding to its receptor in vitro and inhibits tumor growth in vivo. Despite favorable pharmacological activity, the Ang2 mAb preparations were heterogeneous, aggregated rapidly and were poorly expressed. Here, we report the engineering of the antibody variable and constant domains to generate an antibody with reduced propensity to aggregate, enhanced homogeneity, 11°C elevated T(m), 26-fold improved level of expression and retained activity. The engineered molecule, MEDI-3617, is now compatible with the large scale material supply required for clinical trials and is currently being evaluated in Phase 1 in cancer patients. This is the first report to describe the stability engineering of a therapeutic antibody addressing non canonical cysteine residues and the design strategy reported here is generally applicable to other therapeutic antibodies and proteins.


Subject(s)
Antibodies, Monoclonal/genetics , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Protein Engineering/methods , Angiopoietin-2/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Cysteine/metabolism , Hot Temperature , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/therapeutic use , Models, Molecular , Mutagenesis, Site-Directed , Protein Stability
8.
MAbs ; 4(6): 710-23, 2012.
Article in English | MEDLINE | ID: mdl-23007574

ABSTRACT

The c-Met proto-oncogene is a multifunctional receptor tyrosine kinase that is stimulated by its ligand, hepatocyte growth factor (HGF), to induce cell growth, motility and morphogenesis. Dysregulation of c-Met function, through mutational activation or overexpression, has been observed in many types of cancer and is thought to contribute to tumor growth and metastasis by affecting mitogenesis, invasion, and angiogenesis. We identified human monoclonal antibodies that bind to the extracellular domain of c-Met and inhibit tumor growth by interfering with ligand-dependent c-Met activation. We identified antibodies representing four independent epitope classes that inhibited both ligand binding and ligand-dependent activation of c-Met in A549 cells. In cells, the antibodies antagonized c-Met function by blocking receptor activation and by subsequently inducing downregulation of the receptor, translating to phenotypic effects in soft agar growth and tubular morphogenesis assays. Further characterization of the antibodies in vivo revealed significant inhibition of c-Met activity (≥ 80% lasting for 72-96 h) in excised tumors corresponded to tumor growth inhibition in multiple xenograft tumor models. Several of the antibodies identified inhibited the growth of tumors engineered to overexpress human HGF and human c-Met (S114 NIH 3T3) when grown subcutaneously in athymic mice. Furthermore, lead candidate antibody CE-355621 inhibited the growth of U87MG human glioblastoma and GTL-16 gastric xenografts by up to 98%. The findings support published pre-clinical and clinical data indicating that targeting c-Met with human monoclonal antibodies is a promising therapeutic approach for the treatment of cancer.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Proto-Oncogene Proteins c-met/immunology , Animals , Carcinogenesis/drug effects , Carcinogenesis/immunology , Cell Growth Processes/drug effects , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/immunology , Hepatocyte Growth Factor/metabolism , Humans , Immunodominant Epitopes/immunology , Mice , Mice, Nude , Morphogenesis/drug effects , NIH 3T3 Cells , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/genetics , Transgenes/genetics , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 11(8): 1650-60, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22679110

ABSTRACT

The Notch signaling pathway has been implicated in cell fate determination and differentiation in many tissues. Accumulating evidence points toward a pivotal role in blood vessel formation, and the importance of the Delta-like ligand (Dll) 4-Notch1 ligand-receptor interaction has been shown in both physiological and tumor angiogenesis. Disruption of this interaction leads to a reduction in tumor growth as a result of an increase in nonfunctional vasculature leading to poor perfusion of the tumor. MEDI0639 is an investigational human therapeutic antibody that targets Dll4 to inhibit the interaction between Dll4 and Notch1. The antibody cross-reacts to cynomolgus monkey but not mouse species orthologues. In vitro MEDI0639 inhibits the binding of Notch1 to Dll4, interacting via a novel epitope that has not been previously described. Binding to this epitope translates into MEDI0639 reversing Notch1-mediated suppression of human umbilical vein endothelial cell growth in vitro. MEDI0639 administration resulted in stimulation of tubule formation in a three-dimensional (3D) endothelial cell outgrowth assay, a phenotype driven by disruption of the Dll4-Notch signaling axis. In contrast, in a two-dimensional endothelial cell-fibroblast coculture model, MEDI0639 is a potent inhibitor of tubule formation. In vivo, MEDI0639 shows activity in a human endothelial cell angiogenesis assay promoting human vessel formation and reducing the number of vessels with smooth muscle actin-positive mural cells coverage. Collectively, the data show that MEDI0639 is a potent modulator of Dll4-Notch signaling pathway.


Subject(s)
Antibodies, Monoclonal/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Cell Line , Epitope Mapping , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, SCID , Neovascularization, Pathologic , Protein Binding , Receptors, Notch/metabolism , Signal Transduction/drug effects
10.
Cancer Immunol Immunother ; 60(7): 1009-17, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21479995

ABSTRACT

CD40 is a member of the TNF family of receptors that has been shown to play a crucial role in enhancing dendritic cell activity and fostering anti-tumor immune responses. In this study, we demonstrate the in vitro properties and in vivo efficacious activity of the CD40 agonist antibody, CP-870,893. CP-870,893 is a fully human, IgG2 antibody that selectively interacts with CD40 at a site distinct from its ligand-binding region with a KD of 0.4 nM. It enhances the expression of MHC class II, CD54, CD86, and CD23 on human B cells in vitro. CP-870,893 also enhances dendritic cell activity as evidenced by cytokine secretion (IL-12, IL-23, IL-8), the upregulation of CD86 and CD83, and the ability to prime T cells to secrete IFNγ. In SCID-beige mice, a single parenteral injection of CP-870,893 was therapeutically effective against several CD40(pos) human tumors (B-cell lymphoma, breast, colon, and prostate) indicating direct effects on tumor cell survival and/or growth. When mice were co-implanted with human T cells and dendritic cells, the activity of CP-870,893 against CD40(pos) tumors increased, and efficacy was also observed against CD40(neg) and CD40(low) tumors demonstrating the ability of CP-870,893 to enhance anti-tumor immune function in vivo. These studies suggest that CP-870,893 has the potential to be efficacious against a wide range of tumor types through both direct and immune-mediated effects.


Subject(s)
Antibodies, Monoclonal/physiology , B-Lymphocytes/immunology , CD40 Antigens/immunology , Dendritic Cells/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , Animals , Antibodies, Monoclonal, Humanized , B-Lymphocytes/cytology , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/cytology , Flow Cytometry , Humans , Lymphocyte Activation , Mice , Mice, SCID
11.
Mol Cancer Ther ; 9(1): 145-56, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20053776

ABSTRACT

Localized angiopoietin-2 (Ang2) expression has been shown to function as a key regulator of blood vessel remodeling and tumor angiogenesis, making it an attractive candidate for antiangiogenic therapy. A fully human monoclonal antibody (3.19.3) was developed, which may have significant pharmaceutical advantages over synthetic peptide-based approaches in terms of reduced immunogenicity and increased half-life to block Ang2 function. The 3.19.3 antibody potently binds Ang2 with an equilibrium dissociation constant of 86 pmol/L, leading to inhibition of Tie2 receptor phosphorylation in cell-based assays. In preclinical models, 3.19.3 treatment blocked blood vessel formation in Matrigel plug assays and in human tumor xenografts. In vivo studies with 3.19.3 consistently showed broad antitumor activity as a single agent across a panel of diverse subcutaneous and orthotopic xenograft models. Combination studies of 3.19.3 with cytotoxic drugs or anti-vascular endothelial growth factor agents showed significant improvements in antitumor activity over single-agent treatments alone with no apparent evidence of increased toxicity. Initial pharmacokinetic profiling studies in mice and nonhuman primates suggested that 3.19.3 has a predicted human half-life of 10 to 14 days. These studies provide preclinical data for 3.19.3 as a potential new antiangiogenic therapy as a single agent or in combination with chemotherapy or vascular endothelial growth factor inhibitors for the treatment of cancer.


Subject(s)
Angiopoietin-2/immunology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Vascular Endothelial Growth Factors/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibody Specificity/drug effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Collagen/metabolism , Drug Combinations , Humans , Laminin/metabolism , Mice , Neovascularization, Pathologic/drug therapy , Phosphorylation/drug effects , Primates , Protein Binding/drug effects , Proteoglycans/metabolism , Receptor, TIE-2/metabolism
12.
Ann N Y Acad Sci ; 1005: 265-8, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14679073

ABSTRACT

Radioligand binding assays for total and Ig classes of insulin antibodies (IAB) were developed and validated. For each assay, insulin-extracted serum samples were incubated with radiolabeled insulin in the presence and absence of high levels of unlabeled insulin to determine nonspecific binding and total binding, respectively. To measure total IAB, antibody-bound insulin was precipitated with a polyethylene glycol solution, washed, and counted in a gamma-counter. To measure IgG IAB, samples were treated with protein G-Sepharose beads, centrifuged, washed, and counted. For the measurement of IgA, IgE, and IgM IAB, IgG was removed from the samples and treated with anti-IgA, -IgE, or -IgM conjugated to Sepharose beads, centrifuged, washed, and counted. The acid/charcoal extraction of bound and unbound insulin from serum samples was optimized. Specificity and binding capacity of the protein G and antibody-bound beads were evaluated and optimized. The linear region of the total and IgG IAB assays was determined using serum samples containing high levels of insulin antibodies. The limit of quantitation, limit of detection, and precision for all the assays were also determined.


Subject(s)
Autoantibodies/blood , Immunoglobulins/immunology , Insulin/immunology , Antibody Specificity , Humans , Radioligand Assay
13.
J Comp Neurol ; 447(4): 366-80, 2002 Jun 10.
Article in English | MEDLINE | ID: mdl-11992522

ABSTRACT

In the nervous system of the hawkmoth, Manduca sexta, cells expressing the period (per)gene were mapped by in situ hybridization and immunocytochemical methods. Digoxigenin-labeled riboprobes were transcribed from a 1-kb M. sexta per cDNA. Monoclonal anti-PER antibodies were raised to peptide antigens translated from both M. sexta and Drosophila melanogaster per cDNAs. These reagents revealed a widespread distribution of per gene products in M. sexta eyes, optic lobes, brains, and retrocerebral complexes. Labeling for per mRNA was prominent in photoreceptors and in glial cells throughout the brain, and in a cluster of 100-200 neurons adjacent to the accessory medulla of the optic lobes. Daily rhythms of per mRNA levels were detected only in glial cells. PER-like immunoreactivity was observed in nuclei of most neurons and glial cells and in many photoreceptor nuclei. Four neurosecretory cells in the pars lateralis of each brain hemisphere exhibited both nuclear and cytoplasmic staining with anti-PER antibodies. These cells were positively identified as Ia(1) neurosecretory cells that express corazonin immunoreactivity. Anti-corazonin labeled their projections in the brain and their neurohemal endings in the corpora cardiaca and corpora allata. Four pairs of PER-expressing neurosecretory cells previously described in the silkmoth, Anthereae pernyi, are likely to be homologous to these PER/corazonin-expressing Ia(1) cells of M. sexta. Other findings, such as widespread nuclear localization of M. sexta PER and rhythmic expression in glial cells, are reminiscent of the period gene of D. melanogaster, suggesting that some functions of per may be conserved in this lepidopteran species.


Subject(s)
Biological Clocks/genetics , Central Nervous System/metabolism , Circadian Rhythm/genetics , Gene Expression Regulation/physiology , Manduca/genetics , Nuclear Proteins/genetics , RNA, Messenger/metabolism , Animals , Brain/cytology , Brain/metabolism , Central Nervous System/cytology , Drosophila Proteins , Manduca/cytology , Manduca/metabolism , Molecular Probes , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Neurosecretory Systems/cytology , Neurosecretory Systems/metabolism , Nuclear Proteins/metabolism , Optic Lobe, Nonmammalian/cytology , Optic Lobe, Nonmammalian/metabolism , Period Circadian Proteins , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/metabolism , Time Factors
14.
Arch Immunol Ther Exp (Warsz) ; 50(6): 421-9, 2002.
Article in English | MEDLINE | ID: mdl-12546068

ABSTRACT

Peripheral blood lymphocytes from patients with Sezary syndrome (SzS) frequently demonstrate decreased surface expression of transforming growth factor beta receptor II (TGFbetaRII). The mechanism of this low TGFbetaRII expression remains unknown. Because mutations within the poly-A tract of the TGFbetaRII sequence (nucleotides 709-718) were shown to result in diminished TGFbetaRII expression in other types of malignant tumors, we examined the sequence of the TGFbetaRII poly-A tract in two SzS-derived cell lines and in peripheral blood SzS cells from 17 SzS patients and 4 control, healthy individuals using DNA sequencing and single-stranded conformation polymorphism (SSCP) analysis. A standard bidirectional, automated sequence analysis of the RT-PCR-generated cDNA TGFbetaRII fragment showed a heterogenous population of the normal length, 10-, with admixed, shortened, 9-base poly-A stretches. Surprisingly, this mixture was present not only in the cells from 5 SzS patients and 2 SzS cell lines, but also in cells from 2 healthy control individuals. Importantly, the proportion of the shortened, 9-base fragments was markedly reduced or practically eliminated when the procedure was modified by usage of high-fidelity DNA polymerase, labeled primers and/or cloned RT-PCR products, which indicates that the presence of the shortened, 9-base fragments represented a procedural phenomenon rather than a true deletional mutation within an allele of the TGFbetaRII gene. Accordingly, SSCP analysis of genomic DNA did not reveal any mutations within the poly-A tract-containing region. These results indicate that a mechanism different from mutations in the polyadenine tract underlies the diminished TGFbetaRII expression in SzS cells and that the results of an unmodified, direct sequence analysis of homopolymeric base streaches in RT-PCR-derived cDNA should be interpreted with caution.


Subject(s)
DNA, Complementary/chemistry , Mutation , Poly A/genetics , Receptors, Transforming Growth Factor beta/analysis , Receptors, Transforming Growth Factor beta/genetics , Sezary Syndrome/immunology , T-Lymphocytes/chemistry , Base Sequence , Humans , Polymorphism, Single-Stranded Conformational , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...