Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Aging ; 3(8): 931-937, 2023 08.
Article in English | MEDLINE | ID: mdl-37400721

ABSTRACT

Cognitive dysfunction in aging is a major biomedical challenge. Whether treatment with klotho, a longevity factor, could enhance cognition in human-relevant models such as in nonhuman primates is unknown and represents a major knowledge gap in the path to therapeutics. We validated the rhesus form of the klotho protein in mice showing it increased synaptic plasticity and cognition. We then found that a single administration of low-dose, but not high-dose, klotho enhanced memory in aged nonhuman primates. Systemic low-dose klotho treatment may prove therapeutic in aging humans.


Subject(s)
Glucuronidase , Longevity , Mice , Humans , Animals , Aged , Glucuronidase/metabolism , Aging , Cognition , Primates/metabolism
2.
ACS Pharmacol Transl Sci ; 2(6): 485-490, 2019 Dec 13.
Article in English | MEDLINE | ID: mdl-32259079

ABSTRACT

In 2018, the United States Food and Drug Administration (FDA) approved Aimovig (erenumab) for the prevention of migraine. Erenumab is the first FDA approved antibody therapeutic against a G-protein-coupled receptor, the canonical receptor of calcitonin gene related peptide (CGRP-R). A novel, epitope-focused antigen was created to reconstruct the extracellular domains of the CGRP-R in a stable conformation. Successful inoculation of XenoMouse animals and careful screening yielded multiple candidate molecules for high potency and exquisite selectivity toward the CGRP-R over related receptors. These efforts led to the discovery of erenumab which has demonstrated the desired efficacy and safety profiles in multiple clinical studies for the prevention of migraine. The innovation developed in the discovery of erenumab furthers the ability to target G-coupled protein receptors using antibody approaches.

3.
MAbs ; 4(5): 586-91, 2012.
Article in English | MEDLINE | ID: mdl-22820181

ABSTRACT

Peptibodies or peptide-Fc fusions are an attractive alternative therapeutic format to monoclonal antibodies. They consist of biologically active peptides grafted onto an Fc domain. This approach retains certain desirable features of antibodies, notably an increased apparent affinity through the avidity conferred by the dimerization of two Fcs and a long plasma residency time. Peptibodies can be made in E. coli using recombinant technology. The manufacturing process involves fermentation and downstream processing, including refolding and multiple column chromatographic steps, that result in overall yields and quality suitable for commercial development. Romiplostim, marketed under the brand name Nplate®, is the first peptibody to be approved by the United States Food and Drug Administration and the European Medicines Agency and is indicated for the treatment of immune thrombocytopenic purpura. AMG 386, a peptibody antagonist to angiopoietins 1 and 2, is being evaluated in Phase 3 clinical testing in combination with chemotherapy in women with ovarian cancer. AMG 819, a peptibody targeting nerve growth factor for pain has also progressed to clinical trials. These peptibodies illustrate the versatility of the modality.


Subject(s)
Ovarian Neoplasms/therapy , Purpura, Thrombocytopenic, Idiopathic/therapy , Receptors, Fc/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Thrombopoietin/therapeutic use , Clinical Trials as Topic , Female , Humans , Ovarian Neoplasms/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology
4.
Clin Exp Rheumatol ; 30(2): 197-201, 2012.
Article in English | MEDLINE | ID: mdl-22325420

ABSTRACT

OBJECTIVES: AMG623, also known as A-623, is an antagonist of B-cell activating factor (BAFF). The present study was to evaluate the effects of AMG623 on murine models of autoimmune diseases. METHODS: AMG623 was generated through phage library. Inhibitory activities of AMG623 against human and murine BAFF were measured by biacore binding and BAFF-mediated B-cell proliferation assay. Pharmacological effects of AMG623 were studied in BALB/c mice, collagen-induced arthritis model (CIA) and in the NZBxNZW F1 lupus model. RESULTS: AMG623 binds to both soluble and cell surface BAFF. AMG623 blocks both human murine BAFF binding to the receptors. Treatment of AMG623 resulted in B-cell number reduction, and improvement of arthritis and lupus development in mice. CONCLUSIONS: AMG623 is a novel modality of BAFF antagonist. AMG623 is a potential therapeutic agent for the treatment of SLE, rheumatoid arthritis, and other B-cell-mediated autoimmune diseases.


Subject(s)
Arthritis, Experimental/drug therapy , B-Cell Activating Factor/antagonists & inhibitors , B-Lymphocytes/drug effects , Immunologic Factors/pharmacology , Lupus Erythematosus, Systemic/drug therapy , Recombinant Fusion Proteins/pharmacology , Animals , Arthritis, Experimental/immunology , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/metabolism , B-Lymphocytes/immunology , Cell Proliferation/drug effects , Female , HEK293 Cells , Humans , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Time Factors , Transfection
5.
Mol Cancer Ther ; 9(10): 2641-51, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20937592

ABSTRACT

AMG 386 is an investigational first-in-class peptide-Fc fusion protein (peptibody) that inhibits angiogenesis by preventing the interaction of angiopoietin-1 (Ang1) and Ang2 with their receptor, Tie2. Although the therapeutic value of blocking Ang2 has been shown in several models of tumorigenesis and angiogenesis, the potential benefit of Ang1 antagonism is less clear. To investigate the consequences of Ang1 neutralization, we have developed potent and selective peptibodies that inhibit the interaction between Ang1 and its receptor, Tie2. Although selective Ang1 antagonism has no independent effect in models of angiogenesis-associated diseases (cancer and diabetic retinopathy), it induces ovarian atrophy in normal juvenile rats and inhibits ovarian follicular angiogenesis in a hormone-induced ovulation model. Surprisingly, the activity of Ang1 inhibitors seems to be unmasked in some disease models when combined with Ang2 inhibitors, even in the context of concurrent vascular endothelial growth factor inhibition. Dual inhibition of Ang1 and Ang2 using AMG 386 or a combination of Ang1- and Ang2-selective peptibodies cooperatively suppresses tumor xenograft growth and ovarian follicular angiogenesis; however, Ang1 inhibition fails to augment the suppressive effect of Ang2 inhibition on tumor endothelial cell proliferation, corneal angiogenesis, and oxygen-induced retinal angiogenesis. In no case was Ang1 inhibition shown to (a) confer superior activity to Ang2 inhibition or dual Ang1/2 inhibition or (b) antagonize the efficacy of Ang2 inhibition. These results imply that Ang1 plays a context-dependent role in promoting postnatal angiogenesis and that dual Ang1/2 inhibition is superior to selective Ang2 inhibition for suppression of angiogenesis in some postnatal settings.


Subject(s)
Angiopoietin-1/antagonists & inhibitors , Cell Division/drug effects , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/prevention & control , Recombinant Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , Cornea/blood supply , Enzyme-Linked Immunosorbent Assay , Female , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms, Experimental/blood supply , Ovarian Follicle/blood supply , Rats , Rats, Sprague-Dawley
6.
J Pharmacol Exp Ther ; 331(3): 871-81, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19720878

ABSTRACT

Uncontrolled hepatic glucose output (HGO) contributes significantly to the pathological hyperglycemic state of patients with type 2 diabetes. Glucagon, through action on its receptor, stimulates HGO, thereby leading to increased glycemia. Antagonizing the glucagon signaling pathway represents an attractive therapeutic approach for the treatment of type 2 diabetes. We previously reported the generation and characterization of several high-affinity monoclonal antibodies (mAbs) targeting the glucagon receptor (GCGR). In the present study, we demonstrate that a 5-week treatment of diet-induced obese mice with mAb effectively normalized nonfasting blood glucose. Similar treatment also reduced fasting blood glucose without inducing hypoglycemia or other undesirable metabolic perturbations. In addition, no hypoglycemia was found in db/db mice that were treated with a combination of insulin and mAb. Long-term treatment with the mAb caused dose-dependent hyperglucagonemia and minimal to mild alpha-cell hyperplasia in lean mice. There was no evidence of pancreatic alpha-cell neoplastic transformation in mice treated with mAb for as long as 18 weeks. Treatment-induced hyperglucagonemia and alpha-cell hyperplasia were reversible after treatment withdrawal for periods of 4 and 10 weeks, respectively. It is noteworthy that pancreatic beta-cell function was preserved, as demonstrated by improved glucose tolerance throughout the 18-week treatment period. Our studies further support the concept that long-term inhibition of GCGR signaling by a mAb could be an effective approach for controlling diabetic hyperglycemia.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Glucagon-Secreting Cells/pathology , Glucagon/blood , Glucose/metabolism , Hyperglycemia/drug therapy , Hypoglycemic Agents/therapeutic use , Receptors, Glucagon/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Blood Glucose , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Hyperglycemia/blood , Hyperglycemia/metabolism , Hyperplasia , Hypoglycemia/blood , Hypoglycemia/metabolism , Hypoglycemia/prevention & control , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Insulin/administration & dosage , Insulin/therapeutic use , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/metabolism , Tachyphylaxis
7.
J Pharmacol Exp Ther ; 329(1): 102-11, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19129372

ABSTRACT

Antagonizing the glucagon signaling pathway represents an attractive therapeutic approach for reducing excess hepatic glucose production in patients with type 2 diabetes. Despite extensive efforts, there is currently no human therapeutic that directly inhibits the glucagon/glucagon receptor pathway. We undertook a novel approach by generating high-affinity human monoclonal antibodies (mAbs) to the human glucagon receptor (GCGR) that display potent antagonistic activity in vitro and in vivo. A single injection of a lead antibody, mAb B, at 3 mg/kg, normalized blood glucose levels in ob/ob mice for 8 days. In addition, a single injection of mAb B dose-dependently lowered fasting blood glucose levels without inducing hypoglycemia and improved glucose tolerance in normal C57BL/6 mice. In normal cynomolgus monkeys, a single injection improved glucose tolerance while increasing glucagon and active glucagon-like peptide-1 levels. Thus, the anti-GCGR mAb could represent an effective new therapeutic for the treatment of type 2 diabetes.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Glucose/metabolism , Homeostasis/drug effects , Receptors, Glucagon/antagonists & inhibitors , Animals , Blood Glucose/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Endocytosis/drug effects , Flow Cytometry , Glucose Tolerance Test , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Kinetics , Ligands , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
8.
J Bone Miner Res ; 24(4): 578-88, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19049336

ABSTRACT

The development of bone-rebuilding anabolic agents for potential use in the treatment of bone loss conditions, such as osteoporosis, has been a long-standing goal. Genetic studies in humans and mice have shown that the secreted protein sclerostin is a key negative regulator of bone formation, although the magnitude and extent of sclerostin's role in the control of bone formation in the aging skeleton is still unclear. To study this unexplored area of sclerostin biology and to assess the pharmacologic effects of sclerostin inhibition, we used a cell culture model of bone formation to identify a sclerostin neutralizing monoclonal antibody (Scl-AbII) for testing in an aged ovariectomized rat model of postmenopausal osteoporosis. Six-month-old female rats were ovariectomized and left untreated for 1 yr to allow for significant estrogen deficiency-induced bone loss, at which point Scl-AbII was administered for 5 wk. Scl-AbII treatment in these animals had robust anabolic effects, with marked increases in bone formation on trabecular, periosteal, endocortical, and intracortical surfaces. This not only resulted in complete reversal, at several skeletal sites, of the 1 yr of estrogen deficiency-induced bone loss, but also further increased bone mass and bone strength to levels greater than those found in non-ovariectomized control rats. Taken together, these preclinical results establish sclerostin's role as a pivotal negative regulator of bone formation in the aging skeleton and, furthermore, suggest that antibody-mediated inhibition of sclerostin represents a promising new therapeutic approach for the anabolic treatment of bone-related disorders, such as postmenopausal osteoporosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Bone Morphogenetic Proteins/immunology , Bone and Bones/drug effects , Genetic Markers/immunology , Osteogenesis/drug effects , Osteoporosis, Postmenopausal/drug therapy , Animals , Biological Assay , Biomechanical Phenomena , Bone Density/drug effects , Bone and Bones/pathology , Cell Lineage/drug effects , Disease Models, Animal , Female , Femur/drug effects , Femur/pathology , Humans , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/pathology , Mice , Neutralization Tests , Organ Size/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteocalcin/blood , Osteoporosis, Postmenopausal/blood , Osteoporosis, Postmenopausal/pathology , Osteoporosis, Postmenopausal/physiopathology , Ovariectomy , Rats , Rats, Sprague-Dawley , Tibia/drug effects , Tibia/pathology , Tomography, X-Ray Computed
9.
J Bone Miner Res ; 22(10): 1534-47, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17576164

ABSTRACT

UNLABELLED: Skeletal anabolism with PTH is achieved through daily injections that result in brief exposure to the peptide. We hypothesized that similar anabolic effects could be achieved with less frequent but more sustained exposures to PTH. A PTH-Fc fusion protein with a longer half-life than PTH(1-34) increased cortical and cancellous BMD and bone strength with once- or twice-weekly injections. INTRODUCTION: The anabolic effects of PTH are currently achieved with, and thought to require, daily injections that result in brief exposure to the peptide. We hypothesized that less frequent but more sustained exposures to PTH could also be anabolic for bone, provided that serum levels of PTH were not constant. MATERIALS AND METHODS: PTH(1-34) was fused to the Fc fragment of human IgG1 to increase the half-life of PTH. Skeletal anabolism was examined in mice and rats treated once or twice per week with this PTH-Fc fusion protein. RESULTS: PTH-Fc and PTH(1-34) had similar effects on PTH/PTHrP receptor activation, internalization, and signaling in vitro. However, PTH-Fc had a 33-fold longer mean residence time in the circulation of rats compared with that of PTH(1-34). Subcutaneous injection of PTH-Fc once or twice per week resulted in significant increases in bone volume, density, and strength in osteopenic ovariectomized mice and rats. These anabolic effects occurred in association with hypercalcemia and were significantly greater than those achievable with high concentrations of daily PTH(1-34). PTH-Fc also significantly improved cortical bone volume and density under conditions where daily PTH(1-34) did not. Antiresorptive co-therapy with estrogen further enhanced the ability of PTH-Fc to increase bone mass and strength in ovariectomized rats. CONCLUSIONS: These results challenge the notion that brief daily exposure to PTH is essential for its anabolic effects on cortical and cancellous bone. PTH-derived molecules with a sustained circulating half-life may represent a powerful and previously undefined anabolic regimen for cortical and cancellous bone.


Subject(s)
Anabolic Agents/administration & dosage , Anabolic Agents/pharmacology , Bone and Bones/drug effects , Immunoglobulin Fc Fragments/administration & dosage , Parathyroid Hormone/administration & dosage , Parathyroid Hormone/pharmacology , Recombinant Proteins/pharmacology , Aging/physiology , Anabolic Agents/pharmacokinetics , Animals , Arrestins/metabolism , Bone and Bones/metabolism , Cell Line , Cricetinae , Dose-Response Relationship, Drug , Estrogens/pharmacology , Half-Life , Humans , Male , Mice , Ovariectomy , Parathyroid Hormone/pharmacokinetics , Protein Transport , Rats , Receptor, Parathyroid Hormone, Type 1/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Time Factors , beta-Arrestins
10.
Hybridoma (Larchmt) ; 25(3): 115-24, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16796457

ABSTRACT

Keratinocyte growth factor receptor (KGFR) and fibroblast growth factor receptor (FGFR) 2c share identical amino acid sequences, except for a 46-amino acid domain in the extracellular region. Monoclonal antibodies (MAbs) specific to KGFR have not been reported nor are commercially available. In this study, we generated murine MAbs specific to KGFR in non-obese diabetic (NOD) mice using a modified Repeated Immunizations at Multiple Sites (RIMMS) technology. Stable cell lines expressing the full-length human KGFR or FGFR2c were produced to facilitate the identification of KGFR-specific MAbs. Following the initial screening of hybridoma clones with a fluorescence-based, confocal cell detection method and ELISA, KGFR-specific MAbs were selected and confirmed by flow cytometry and Western blot analyses. Antagonistic MAbs were identified using a cell-based functional assay. These KGFR MAbs will be important reagents for studying the biological function and tissue distribution of this receptor in normal and pathological conditions.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Receptor, Fibroblast Growth Factor, Type 2/immunology , Amino Acid Sequence , Animals , Antibody Specificity , Blotting, Western , CHO Cells , Cell Line , Cricetinae , Cricetulus , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Molecular Sequence Data
11.
Cancer Cell ; 6(5): 507-16, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15542434

ABSTRACT

Angiopoietin-2 (Ang2) exhibits broad expression in the remodeling vasculature of human tumors but very limited expression in normal tissues, making it an attractive candidate target for antiangiogenic cancer therapy. To investigate the functional consequences of blocking Ang2 activity, we generated antibodies and peptide-Fc fusion proteins that potently and selectively neutralize the interaction between Ang2 and its receptor, Tie2. Systemic treatment of tumor-bearing mice with these Ang2-blocking agents resulted in tumor stasis, followed by elimination of all measurable tumor in a subset of animals. These effects were accompanied by reduced endothelial cell proliferation, consistent with an antiangiogenic therapeutic mechanism. Anti-Ang2 therapy also prevented VEGF-stimulated neovascularization in a rat corneal model of angiogenesis. These results imply that specific Ang2 inhibition may represent an effective antiangiogenic strategy for treating patients with solid tumors.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Angiopoietin-2/immunology , Antibodies/pharmacology , Neoplasms/blood supply , Neovascularization, Pathologic/prevention & control , Animals , Cell Proliferation/drug effects , Cornea/blood supply , Endothelial Cells/drug effects , Female , Mice , Mice, Nude , Neoplasm Transplantation , Neutralization Tests , Receptors, Fc , Recombinant Fusion Proteins/pharmacology , Tumor Cells, Cultured
12.
Clin Cancer Res ; 9(10 Pt 2): 3982S-90S, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14506197

ABSTRACT

PURPOSE: Epratuzumab is a novel humanized antihuman CD22 IgG1 antibody that has recently shown promising clinical activity, both as a single agent and in combination with rituximab, in patients with non-Hodgkin's lymphomas (NHL). In an attempt to better understand the mode of action of epratuzumab, the antibody was tested in vitro in a variety of cell-based assays similar to those used to evaluate the biological activity of other therapeutic monoclonal antibodies, including rituximab. In this report, we present epratuzumab activities as they relate to binding, signaling, and internalization of the receptor CD22. METHODS: Chinese hamster ovary-expressed CD22 extracellular domain was used to measure epratuzumab affinity on Biacore. CD22 receptor density and internalization rate were measured indirectly using a monovalently labeled, noncompeting (with epratuzumab) anti-CD22 antibody on Burkitt lymphoma cell lines, primary B cells derived from fresh tonsils, and B cells separated from peripheral blood samples obtained from patients with chronic lymphocytic leukemia or healthy volunteers. Epratuzumab-induced CD22 phosphorylation was measured by immunoprecipitation/Western blot and compared with that induced by anti-IgM stimulation. RESULTS: Epratuzumab binds to CD22-extracellular domain, with an affinity of K(D) = 0.7 nM. Binding of epratuzumab to B cell lines, or primary B cells from healthy individuals and patients with NHL, results in rapid internalization of the CD22/antibody complex. Internalization appears to be faster at early time points in cell lines than in primary B cells and NHL patient-derived B cells, but the maximum internalization reached is comparable for all B cell populations after several hours of treatment and appears to reach saturation at antibody concentrations of 1-5 micro g/ml. Finally, epratuzumab binding results in modest but significant CD22 phosphorylation. CONCLUSIONS: Epratuzumab represents an excellent anti-CD22 ligating agent, highly efficacious in inducing CD22 internalization, and can induce phosphorylation. Although we cannot unequivocally demonstrate here that epratuzumab-induced internalization and signaling of CD22 directly contribute to its therapeutic efficacy, these properties are the fundamental characteristics of the target CD22 and its interaction with epratuzumab. Similar results were observed when epratuzumab was tested in vitro on Burkitt B cell lines as well as on primary normal B cells and neoplastic B cells separated from fresh peripheral blood samples from patients with chronic lymphocytic leukemia.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/biosynthesis , Antigens, Differentiation, B-Lymphocyte/biosynthesis , Cell Adhesion Molecules , Lectins/biosynthesis , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antigens, CD/genetics , Antigens, CD19/biosynthesis , Antigens, CD20/biosynthesis , Antigens, Differentiation, B-Lymphocyte/genetics , Blotting, Western , CHO Cells , Cell Line , Cell Line, Tumor , Cloning, Molecular , Cricetinae , Humans , Immunoglobulin M/chemistry , In Vitro Techniques , Kinetics , Lectins/genetics , Microscopy, Confocal , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Rituximab , Sialic Acid Binding Ig-like Lectin 2 , Time Factors
13.
J Immunol ; 169(3): 1151-8, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12133934

ABSTRACT

NF-kappa B-inducing kinase (NIK) is involved in lymphoid organogenesis in mice through lymphotoxin-beta receptor signaling. To clarify the roles of NIK in T cell activation through TCR/CD3 and costimulation pathways, we have studied the function of T cells from aly mice, a strain with mutant NIK. NIK mutant T cells showed impaired proliferation and IL-2 production in response to anti-CD3 stimulation, and these effects were caused by impaired NF-kappa B activity in both mature and immature T cells; the impaired NF-kappa B activity in mature T cells was also associated with the failure of maintenance of activated NF-kappa B. In contrast, responses to costimulatory signals were largely retained in aly mice, suggesting that NIK is not uniquely coupled to the costimulatory pathways. When NIK mutant T cells were stimulated in the presence of a protein kinase C (PKC) inhibitor, proliferative responses were abrogated more severely than in control mice, suggesting that both NIK and PKC control T cell activation in a cooperative manner. We also demonstrated that NIK and PKC are involved in distinct NF-kappa B activation pathways downstream of TCR/CD3. These results suggest critical roles for NIK in setting the threshold for T cell activation, and partly account for the immunodeficiency in aly mice.


Subject(s)
Lymphocyte Activation , Protein Serine-Threonine Kinases/physiology , Receptor-CD3 Complex, Antigen, T-Cell/physiology , T-Lymphocytes/immunology , Animals , B-Cell Lymphoma 3 Protein , I-kappa B Kinase , Interleukin-2/physiology , Mice , NF-kappa B/metabolism , Protein Kinase C/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/physiology , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...