Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
3.
Mol Cancer Ther ; 19(2): 513-524, 2020 02.
Article in English | MEDLINE | ID: mdl-31871267

ABSTRACT

Cytotoxic T-lymphocyte antigen 4 (CTLA4)-FasL, a homo-hexameric signal converter protein, is capable of inducing robust apoptosis in malignant cells of the B-cell lineage expressing its cognate B7 and Fas targets, while sparing nonmalignant ones. This fusion protein's striking proapoptotic efficacy stems from its complementary abilities to coordinately activate apoptotic signals and abrogate antiapoptotic ones. A limiting factor in translating FasL or Fas receptor agonists into the clinic has been lethal hepatotoxicity. Here, we establish CTLA4-FasL's in vivo efficacy in multiple murine and xenograft models, for both systemic and subcutaneous tumors. Significantly, good laboratory practice (GLP) toxicology studies in mice indicate that CTLA4-FasL given repeatedly at doses up to five times the effective dose was well-tolerated and resulted in no significant adverse events. An equivalent single dose of CTLA4-FasL administered to nonhuman primates was also well-tolerated, albeit with a moderate dose-dependent leukopenia that was completely reversible. Interestingly, monkey peripheral blood mononuclear cells were more sensitive to CTLA4-FasL-induced apoptosis when tested in vitro. In both species, there was short-term elevation in serum levels of IL6, IL2, and IFNγ, although this was not associated with clinical signs of proinflammatory cytokine release, and further, this cytokine elevation could be completely prevented by dexamethasone premedication. Liver toxicity was not observed in either species, as confirmed by serum liver enzyme levels and histopathologic assessment. In conclusion, CTLA4-FasL emerges from animal model studies as an effective and safe agent for targeted FasL-mediated treatment of B7-expressing aggressive B-cell lymphomas.


Subject(s)
CTLA-4 Antigen/administration & dosage , Fas Ligand Protein/administration & dosage , Recombinant Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , CTLA-4 Antigen/immunology , Fas Ligand Protein/adverse effects , Fas Ligand Protein/immunology , Fas Ligand Protein/pharmacokinetics , Female , Humans , Jurkat Cells , Macaca fascicularis , Mice , Mice, Inbred BALB C , Mice, Nude , Primates , Random Allocation , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Xenograft Model Antitumor Assays
4.
Mol Cancer Res ; 14(7): 599-611, 2016 07.
Article in English | MEDLINE | ID: mdl-27053682

ABSTRACT

UNLABELLED: Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal cancers, in part, due to resistance to both conventional and targeted therapeutics. TRAIL directly induces apoptosis through engagement of cell surface Death Receptors (DR4 and DR5), and has been explored as a molecular target for cancer treatment. Clinical trials with recombinant TRAIL and DR-targeting agents, however, have failed to show overall positive outcomes. Herein, we identify a novel TRAIL resistance mechanism governed by Hu antigen R (HuR, ELAV1), a stress-response protein abundant and functional in PDA cells. Exogenous HuR overexpression in TRAIL-sensitive PDA cell lines increases TRAIL resistance whereas silencing HuR in TRAIL-resistant PDA cells, by siRNA oligo-transfection, decreases TRAIL resistance. PDA cell exposure to soluble TRAIL induces HuR translocation from the nucleus to the cytoplasm. Furthermore, it is demonstrated that HuR interacts with the 3'-untranslated region (UTR) of DR4 mRNA. Pre-treatment of PDA cells with MS-444 (Novartis), an established small molecule inhibitor of HuR, substantially increased DR4 and DR5 cell surface levels and enhanced TRAIL sensitivity, further validating HuR's role in affecting TRAIL apoptotic resistance. NanoString analyses on the transcriptome of TRAIL-exposed PDA cells identified global HuR-mediated increases in antiapoptotic processes. Taken together, these data extend HuR's role as a key regulator of TRAIL-induced apoptosis. IMPLICATIONS: Discovery of an important new HuR-mediated TRAIL resistance mechanism suggests that tumor-targeted HuR inhibition increases sensitivity to TRAIL-based therapeutics and supports their re-evaluation as an effective treatment for PDA patients. Mol Cancer Res; 14(7); 599-611. ©2016 AACR.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , ELAV-Like Protein 1/metabolism , Pancreatic Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Recombinant Proteins/pharmacology , Transfection
5.
Acad Med ; 90(4): 505-10, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25629944

ABSTRACT

PURPOSE: To test the hypotheses that medical students recognized by peers as the most positive social influencers would score (1) high on measures of engaging personality attributes that are conducive to relationship building (empathy, sociability, activity, self-esteem), and (2) low on disengaging personality attributes that are detrimental to interpersonal relationships (loneliness, neuroticism, aggression-hostility, impulsive sensation seeking). METHOD: The study included 666 Jefferson Medical College students who graduated in 2011-2013. Students used a peer nomination instrument to identify classmates who had a positive influence on their professional and personal development. At matriculation, these students had completed a survey that included the Jefferson Scale of Empathy and Zuckerman-Kuhlman Personality Questionnaire short form and abridged versions of the Rosenberg Self-Esteem Scale and UCLA Loneliness Scale. In multivariate analyses of variance, the method of contrasted groups was used to compare the personality attributes of students nominated most frequently by their peers as positive influencers (top influencers [top 25% in their class distribution], n = 176) with those of students nominated least frequently (bottom influencers [bottom 25%], n = 171). RESULTS: The top influencers scored significantly higher on empathy, sociability, and activity and significantly lower on loneliness compared with the bottom influencers. However, the effect size estimates of the differences were moderate at best. CONCLUSIONS: The research hypotheses were partially confirmed. Positive social influencers appear to possess personality attributes conducive to relationship building, which is an important feature of effective leadership. The findings have implications for identifying and training potential leaders in medicine.


Subject(s)
Empathy , Leadership , Personality , Students, Medical/psychology , Aggression , Anxiety Disorders , Female , Hostility , Humans , Impulsive Behavior , Interpersonal Relations , Loneliness , Male , Neuroticism , Philadelphia , Self Concept , Social Skills
6.
Med Teach ; 37(7): 677-683, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25155553

ABSTRACT

BACKGROUND: Previous research has paid little to no attention towards exploring methods of identifying existing medical student leaders. AIM: Focusing on the role of influence and employing the tenets of the engaging leadership model, this study examines demographic and academic performance-related differences of positive influencers and if students who have been peer-identified as positive influencers also demonstrate high levels of genuine concern for others. METHODS: Three separate fourth-year classes were asked to designate classmates that had significant positive influences on their professional and personal development. The top 10% of those students receiving positive influence nominations were compared with the other students on demographics, academic performance, and genuine concern for others. RESULTS: Besides age, no demographic differences were found between positive influencers and other students. High positive influencers were not found to have higher standardized exam scores but did receive significantly higher clinical clerkship ratings. High positive influencers were found to possess a higher degree of genuine concern for others. CONCLUSION: The findings lend support to (a) utilizing the engaging model to explore leaders and leadership within medical education, (b) this particular method of identifying existing medical student leaders, and

7.
PLoS One ; 8(10): e77050, 2013.
Article in English | MEDLINE | ID: mdl-24130833

ABSTRACT

BACKGROUND: New strategies for the treatment of hepatocellular carcinoma (HCC) are needed, given that currently available chemotherapeutics are inefficient. Since tumor growth reflects the net balance between pro-proliferative and death signaling, agents shifting the equilibrium toward the latter are of considerable interest. The TWEAK:Fn14 signaling axis promotes tumor cell proliferation and tumor angiogenesis, while TRAIL:TRAIL-receptor (TRAIL-R) interactions selectively induce apoptosis in malignant cells. Fn14•TRAIL, a fusion protein bridging these two pathways, has the potential to inhibit tumor growth, by interfering with TWEAK:Fn14 signaling, while at the same time enforcing TRAIL:TRAIL-R-mediated apoptosis. Consequently, Fn14•TRAIL's capacity to inhibit HCC growth was tested. RESULTS: Fn14•TRAIL induced robust apoptosis of multiple HCC cell lines, while sparing non-malignant hepatocyte cell lines. Differential susceptibility to this agent did not correlate with expression levels of TRAIL, TRAIL-R, TWEAK and Fn14 by these lines. Fn14•TRAIL was more potent than soluble TRAIL, soluble Fn14, or a combination of the two. The requirement of both of Fn14•TRAIL's molecular domains for function was established using blocking antibodies directed against each of them. Subcutaneous injection of Fn14•TRAIL abrogated HCC growth in a xenograft model, and was well tolerated by the mice. CONCLUSIONS: In this study, Fn14•TRAIL, a multifunctional fusion protein originally designed to treat autoimmunity, was shown to inhibit the growth of HCC, both in vitro and in vivo. The demonstration of this fusion protein's potent anti-tumor activity suggests that simultaneous targeting of two signaling axes by a single fusion can serve as a basis for highly effective anti-cancer therapies.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Receptors, Tumor Necrosis Factor/genetics , Recombinant Fusion Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokine TWEAK , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Signal Transduction/drug effects , Solubility , TNF-Related Apoptosis-Inducing Ligand/chemistry , TWEAK Receptor , Tumor Necrosis Factors/genetics , Xenograft Model Antitumor Assays
8.
Cell Cycle ; 10(24): 4208-16, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22033146

ABSTRACT

It is now widely recognized that the tumor microenvironment promotes cancer cell growth and metastasis via changes in cytokine secretion and extracellular matrix remodeling. However, the role of tumor stromal cells in providing energy for epithelial cancer cell growth is a newly emerging paradigm. For example, we and others have recently proposed that tumor growth and metastasis is related to an energy imbalance. Host cells produce energy-rich nutrients via catabolism (through autophagy, mitophagy, and aerobic glycolysis), which are then transferred to cancer cells to fuel anabolic tumor growth. Stromal cell-derived L-lactate is taken up by cancer cells and is used for mitochondrial oxidative phosphorylation (OXPHOS) to produce ATP efficiently. However, "parasitic" energy transfer may be a more generalized mechanism in cancer biology than previously appreciated. Two recent papers in Science and Nature Medicine now show that lipolysis in host tissues also fuels tumor growth. These studies demonstrate that free fatty acids produced by host cell lipolysis are re-used via beta-oxidation (beta-OX) in cancer cell mitochondria. Thus, stromal catabolites (such as lactate, ketones, glutamine and free fatty acids) promote tumor growth by acting as high-energy onco-metabolites. As such, host catabolism, via autophagy, mitophagy and lipolysis, may explain the pathogenesis of cancer-associated cachexia and provides exciting new druggable targets for novel therapeutic interventions. Taken together, these findings also suggest that tumor cells promote their own growth and survival by behaving as a "parasitic organism." Hence, we propose the term "Parasitic Cancer Metabolism" to describe this type of metabolic coupling in tumors. Targeting tumor cell mitochondria (OXPHOS and beta-OX) would effectively uncouple tumor cells from their hosts, leading to their acute starvation. In this context, we discuss new evidence that high-energy onco-metabolites (produced by the stroma) can confer drug resistance. Importantly, this metabolic chemo-resistance is reversed by blocking OXPHOS in cancer cell mitochondria with drugs like Metformin, a mitochondrial "poison." In summary, parasitic cancer metabolism is achieved architecturally by dividing tumor tissue into at least two well-defined opposing "metabolic compartments:" catabolic and anabolic.


Subject(s)
Cachexia/metabolism , Energy Metabolism/physiology , Metabolic Networks and Pathways/physiology , Mitochondria/metabolism , Models, Biological , Neoplasms/metabolism , Stromal Cells/metabolism , Cachexia/etiology , Fibroblasts/physiology , Glycolysis/physiology , Lipolysis/physiology , Neoplasms/complications , Oxidative Phosphorylation
9.
Am J Pathol ; 177(6): 3159-68, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21088216

ABSTRACT

Evolution of apoptosis resistance in both lymphoma and leukemia cells is well documented, and induction of apoptosis in malignant cells is a major goal of cancer therapy. Up-regulation of anti-apoptotic signals is one of the mechanisms whereby resistance to apoptosis emerges. We have previously described the fusion proteins CD40·FasL and CTLA-4·FasL, which are formed from two functional membrane proteins and induce apoptosis of activated T cells. The present study explores the potential use of CD40·FasL and CTLA-4·FasL for the killing of malignant cells of lymphatic origin. Using malignant B and T cell lines that differ in surface expression of costimulatory molecules, we found that CTLA-4·FasL induces effective apoptosis of cells expressing CD95 and activates caspases 3, 8, and 9. Only B7-expressing B cells responded to CTLA-4·FasL with rapid abrogation of cFLIP expression. CD40·FasL effectively killed only the T cells that express high levels of CD40L in addition to CD95. In these cells, CD40·FasL significantly diminished cFLIP expression. Importantly, each of the fusion proteins is more potent than its respective components parts, alone or in combination. Thus, the proteins with their two functional ends deliver a pro-apoptotic signal and, in parallel, inhibit an anti-apoptotic signal, thus optimizing the wanted, death-inducing effect. Therefore, these proteins emerge as promising agents to be used for targeted and specific tumor cell killing.


Subject(s)
Antigens, CD/pharmacology , Apoptosis/drug effects , CD40 Antigens/pharmacology , Fas Ligand Protein/pharmacology , Neoplasms/pathology , Recombinant Fusion Proteins/pharmacology , Antigens, CD/genetics , CD40 Antigens/genetics , CTLA-4 Antigen , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Fas Ligand Protein/genetics , Humans , Jurkat Cells , Molecular Targeted Therapy , Neoplasms/drug therapy , Recombinant Fusion Proteins/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
10.
PLoS One ; 5(9): e12868, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20886106

ABSTRACT

BACKGROUND: T-helper polarization of naïve T cells is determined by a complex mechanism that involves many factors, eventually leading to activation of Th1, Th2, or Th17 responses or alternatively the generation of regulatory T cells. Placental Protein 14 (PP14) is a 28 kDa glycoprotein highly secreted in early pregnancy that is able to desensitize T cell receptor (TCR) signaling and modulate T cell activation. METHODOLOGY/PRINCIPAL FINDINGS: Prolonged antigen-specific stimulation of T cells in the presence of PP14 resulted in an impaired secretion of IFN-γ, IL-5 and IL-17 upon restimulation, although the cells proliferated and expressed activation markers. Furthermore, the generation of regulatory CD4(+)CD25(high)Foxp3(+) T cells was induced in the presence of PP14, in both antigen-specific as well as polyclonal stimulation. In accordance with previous reports, we found that the induction of FoxP3 expression by PP14 is accompanied by down regulation of the PI3K-mTOR signaling pathway. CONCLUSIONS/SIGNIFICANCE: These data suggest that PP14 arrests T cells in a unique activated state that is not accompanied with the acquisition of effector function, together with promoting the generation of regulatory T cells. Taken together, our results may elucidate the role of PP14 in supporting immune tolerance in pregnancy by reducing T cell effector functions along with augmenting Treg differentiation.


Subject(s)
Cell Differentiation , Forkhead Transcription Factors/genetics , Glycoproteins/immunology , Pregnancy Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Adult , Cells, Cultured , Female , Forkhead Transcription Factors/immunology , Gene Expression , Glycodelin , Glycoproteins/genetics , Humans , Lymphocyte Activation , Male , Middle Aged , Pregnancy Proteins/genetics , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Young Adult
11.
Am J Pathol ; 174(2): 460-74, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19147815

ABSTRACT

Hallmarks of the pathogenesis of autoimmune encephalomyelitis include perivascular infiltration of inflammatory cells into the central nervous system, multifocal demyelination in the brain and spinal cord, and focal neuronal degeneration. Optimal treatment of this complex disease will ultimately call for agents that target the spectrum of underlying pathogenic processes. In the present study, Fn14-TRAIL is introduced as a unique immunotherapeutic fusion protein that is designed to exchange and redirect intercellular signals within inflammatory cell networks, and, in so doing, to impact multiple pathogenic events and yield a net anti-inflammatory effect. In this soluble protein product, a Fn14 receptor component (capable of blocking the pro-inflammatory TWEAK ligand) is fused to a TRAIL ligand (capable of inhibiting activated, pathogenic T cells). Sustained Fn14-TRAIL expression was obtained in vivo using a transposon-based eukaryotic expression vector. Fn14-TRAIL expression effectively prevented chronic, nonremitting, paralytic disease in myelin oligodendrocyte glycoprotein-challenged C57BL/6 mice. Disease suppression in this model was reflected by decreases in the clinical score, disease incidence, nervous tissue inflammation, and Th1, Th2, and Th17 cytokine responses. Significantly, the therapeutic efficacy of Fn14-TRAIL could not be recapitulated simply by administering its component parts (Fn14 and TRAIL) as soluble agents, either alone or in combination. Its functional pleiotropism was manifest in its additional ability to attenuate the enhanced permeability of the blood-brain barrier that typically accompanies autoimmune encephalomyelitis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunologic Factors/therapeutic use , Receptors, Tumor Necrosis Factor/therapeutic use , Recombinant Fusion Proteins/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Animals , Brain/drug effects , Brain/pathology , CHO Cells , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunologic Factors/immunology , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor/immunology , Recombinant Fusion Proteins/immunology , Spinal Cord/drug effects , Spinal Cord/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , TWEAK Receptor
12.
Int Immunol ; 20(4): 471-83, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18272926

ABSTRACT

CTLA-4 x Ig was originally designed as an immunosuppressive agent capable of interfering with the co-stimulation of T cells. In the present study, we demonstrate that CTLA-4 x Ig, in combination with TCR ligation, has the additional capacity to convert naive CD4+CD25- T cells into Foxp3+ regulatory T (T(reg)) cells, as well as to expand their numbers. The CD4+CD25+Foxp3+ T(reg) generated by CTLA-4 x Ig treatment in vitro potently suppress effector T cells. Extending this in vivo, we show that systemic administration of CTLA-4 x Ig increases the percentage of CD4+CD25(hi)Foxp3+ cells within mixed lymphocyte reaction-induced murine lymph nodes. Significantly, the in vitro conversion of naive CD4+CD25- T cells into T(reg) cells is antigen-presenting cell (APC) dependent. This finding, together with the further observation that this conversion can also be driven in vitro by an antibody that engages B7-2 ligand, suggests that CTLA-4 x Ig-driven T(reg) induction may be predicated upon active CTLA-4 x Ig to B7-2 signaling within APC, which elicits from them T(reg)-inducing potential. These findings extend CTLA-4 x Ig's functional repertoire, and at the same time, reinforce the concept that T cell anergy and active suppression are not entirely distinct processes and may be linked by some common molecular triggers.


Subject(s)
Antigens, CD/pharmacology , Immunoglobulins/pharmacology , Interleukin-2 Receptor alpha Subunit/biosynthesis , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies/blood , Antigen-Antibody Reactions , CD4 Antigens/biosynthesis , CTLA-4 Antigen , Cell Proliferation/drug effects , Cells, Cultured , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , T-Lymphocytes, Regulatory/drug effects
13.
Nat Immunol ; 9(2): 194-202, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18157133

ABSTRACT

The transcription factor Foxp3 is involved in the differentiation, function and survival of CD4+CD25+ regulatory T (T(reg)) cells. Details of the mechanism underlying the induction of Foxp3 expression remain unknown, because studies of the transcriptional regulation of the Foxp3 gene are limited by the small number of T(reg) cells in mononuclear cell populations. Here we have generated a model system for analyzing Foxp3 induction and, by using this system with primary T cells, we have identified an enhancer element in this gene. The transcription factors Smad3 and NFAT are required for activity of this Foxp3 enhancer, and both factors are essential for histone acetylation in the enhancer region and induction of Foxp3. These biochemical properties that define Foxp3 expression explain many of the effects of transforming growth factor-beta on the function of Foxp3+ T(reg) cells.


Subject(s)
Enhancer Elements, Genetic , Forkhead Transcription Factors/genetics , Gene Expression Regulation , NFATC Transcription Factors/metabolism , Smad3 Protein/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Base Sequence , Cell Line, Tumor , Mice , Molecular Sequence Data
14.
J Immunol ; 179(11): 7287-94, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18025171

ABSTRACT

The fusion protein CTLA-4 . FasL, a paradigmatic "trans signal converter protein", can attach to APC surfaces and in effect convert B7-activating costimulator signals into inhibitory Fas receptor-generated signals. The present study investigates CTLA-4 . FasL's mechanism of action. A combination of p27(kip) and proliferating cell nuclear Ag Western blot and propidium iodide flow cytometric analysis showed no CTLA-4 . FasL effect on cell cycle entry and progression, pointing away from the kind of classical anergy associated with CTLA-4 . Ig. Significantly, CTLA-4 . FasL elicited apoptosis (as detected by annexin-V/propidium iodide costaining) as early as 24 h after T cell activation, suggesting that some coordinate signaling might be capacitating the Fas receptor. Significantly, CTLA-4 . FasL, but not CTLA-4 . Ig, anti-Fas mAb, or the two in combination, abrogated the usual increase in expression of the anti-apototic protein, cFLIP. Furthermore, activation of caspases 8 and 3 were not affected by CTLA-4 . FasL. These findings suggest a model for CTLA-4 . FasL action wherein there is coordinate triggering of a death receptor and suppression of a proapoptotic protein.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation/immunology , Apoptosis/immunology , Fas Ligand Protein/immunology , Recombinant Fusion Proteins/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antigens, CD/drug effects , Antigens, Differentiation/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/immunology , CTLA-4 Antigen , Cell Cycle/immunology , Cell Proliferation/drug effects , Cells, Cultured , Fas Ligand Protein/antagonists & inhibitors , Female , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/antagonists & inhibitors , Up-Regulation/immunology
15.
J Immunol ; 179(3): 1760-7, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641042

ABSTRACT

OX40 is a member of the TNFR superfamily (CD134; TNFRSF4) that is expressed on activated T cells and regulates T cell-mediated immune responses. In this study, we have examined the regulation of OX40 gene expression in T cells. Low-level OX40 mRNA expression was detected in both resting T cells and the nonactivated EL4 T cell line, and was up-regulated in both types of T cells upon activation with anti-CD3 Ab. We have shown in this study that basal OX40 promoter activity is regulated by constitutively expressed Sp1/Sp3 and YY1 transcription factors. NF-kappaB (p50 and p65) also binds to the OX40 promoter region, but the level of direct enhancement of the OX40 promoter activity by this transcription factor is not sufficient to account for the observed up-regulation of OX40 mRNA expression associated with activation. We have detected by chromatin immunoprecipitation that histone H4 molecules in the OX40 promoter region are highly acetylated by activation and NF-kappaB binds to the OX40 promoter in vivo. These findings suggest that OX40 gene expression is regulated by chromatin remodeling, and that NF-kappaB might be involved in initiation of chromatin remodeling in the OX40 promoter region in activated T cells. CD4(+)CD25(+) regulatory T (Treg) cells also express OX40 at high levels, and signaling through this receptor can neutralize suppressive activity of this Treg cell. In CD4(+)CD25(+) Treg cells, histone H4 molecules in the OX40 promoter region are also highly acetylated, even in the absence of in vitro activation.


Subject(s)
Chromatin Assembly and Disassembly/immunology , NF-kappa B p50 Subunit/metabolism , Promoter Regions, Genetic/immunology , Receptors, OX40/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Up-Regulation/immunology , YY1 Transcription Factor/metabolism , Animals , Binding Sites/genetics , Binding Sites/immunology , CD3 Complex/immunology , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , Immune Sera/pharmacology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/metabolism , Mice , RNA, Messenger/biosynthesis , Receptors, OX40/biosynthesis , Receptors, OX40/metabolism , Sp1 Transcription Factor/physiology , Sp3 Transcription Factor/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription Factor RelA/metabolism , Up-Regulation/genetics , YY1 Transcription Factor/physiology
16.
Glycobiology ; 16(3): 173-83, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16269626

ABSTRACT

Placental protein 14 (PP14; glycodelin) is a pregnancy-associated immunoregulatory protein that is known to inhibit T cells via T-cell receptor desensitization. The recent demonstration of PP14 as lectin has provided insight into how it may mediate its CD45 glycoprotein-dependent T-cell inhibition. In this study, we have investigated PP14's lectin-binding properties in detail. Significantly, PP14 reacts with N-acetyllactosamine (LacNAc) as was also found for members of the galectin family, such as the potent immunoregulatory protein, galectin-1. However, in contrast to galectin-1, PP14's binding is significantly enhanced by alpha2,6-sialylation and also by the presence of cations. This was demonstrated by preferential binding to fetuin as compared with its desialylated variant asialofetuin (ASF) and by using free alpha2,6- versus alpha2,3-sialylated forms of LacNAc in competitive inhibition and direct solid-phase binding assays. Interestingly, from immunological point of view, PP14 also binds differentially to CD45 isoforms known to differ in their degree of sialylation. PP14 preferentially inhibits CD45RA+, as compared with CD45RO+ T cells, and preferentially co-capped this variant CD45 on the T-cell surface. Finally, we demonstrate that PP14 promotes CD45 dimerization and clustering, a phenomenon that may regulate CD45 activity.


Subject(s)
ABO Blood-Group System/immunology , Calcium/pharmacology , Glycoproteins/metabolism , Lectins/metabolism , Leukocyte Common Antigens/metabolism , Lymphocytes/immunology , N-Acetylneuraminic Acid/metabolism , Pregnancy Proteins/metabolism , Calcium/chemistry , Cations, Divalent/chemistry , Cells, Cultured , Dimerization , Female , Glycodelin , Glycoproteins/chemistry , Humans , Lectins/chemistry , Leukocyte Common Antigens/immunology , Lymphocytes/chemistry , Lymphocytes/drug effects , Lymphocytes/metabolism , Pregnancy , Pregnancy Proteins/chemistry , Protein Binding/drug effects , Protein Isoforms/immunology , Protein Isoforms/metabolism
17.
Lab Invest ; 85(9): 1058-64, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16056243

ABSTRACT

Academic Pathology is a key player in human molecular science and in the powerful initiatives of the National Institutes of Health. Pathologists generate data crucial to virtually every molecular study of human tissue, and have the necessary skills and authority to oversee processing of human tissues for research analysis. We advocate that Academic Pathology is optimally positioned to drive the molecular revolution in study of human disease, through human tissue collection, analysis, and databasing. This can be achieved through playing a major role in human tissue procurement and management; establishing high-quality 'Pathology Resource Laboratories'; providing the scientific expertise for pathology data sharing; and recruiting and training physician scientists. Pathology should position itself to be the local institutional driver of technology implementation and development, by operating the resource laboratories, providing the expertise for technical and conceptual design of research projects, maintaining the databases that link molecular and morphological information on human tissues with the requisite clinical databases, providing education and mentorship of technology users, and nurturing new research through the development of preliminary data. We also consider that outstanding pathology journals are available for the publication of research emanating from such studies, to the benefit of the pathology profession as an academic enterprise. It is our earnest hope that Academic Pathology can play a leading role in the remarkable advances to be made as the 21st century unfolds.


Subject(s)
Biomedical Research , Pathology , Humans , Laboratories
18.
J Immunol ; 173(9): 5524-30, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15494501

ABSTRACT

The potency of TCR signaling during primary CD4+ T cell activation influences initial cytokine expression patterns and subsequent polarization toward either Th1 or Th2 subsets. In this study, we demonstrate that the T cell inhibitor placental protein 14 (PP14; glycodelin) preferentially inhibits Th1 cytokine responses and chemokine expression when present during ex vivo priming of CD4+ T cells. PP14 synergizes with exogenously added IL-4 in skewing T cell responses. Significantly, PP14 impairs the down-regulation of GATA-3 transcriptional regulator expression that normally accompanies T cell activation, which is a prerequisite for Th1 development. Taken together, these data document for the first time the ability of PP14 to skew Th responses.


Subject(s)
Cytokines/biosynthesis , Glycoproteins/physiology , Pregnancy Proteins/physiology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Amniotic Fluid/immunology , Amniotic Fluid/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , Down-Regulation/genetics , Down-Regulation/immunology , Enterotoxins/pharmacology , GATA3 Transcription Factor , Glycodelin , Glycoproteins/deficiency , Glycoproteins/genetics , Humans , Pregnancy Proteins/deficiency , Pregnancy Proteins/genetics , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Trans-Activators/antagonists & inhibitors , Trans-Activators/biosynthesis
19.
Cell Immunol ; 232(1-2): 144-56, 2004.
Article in English | MEDLINE | ID: mdl-15882859

ABSTRACT

A successful pregnancy can only occur when the maternal immune system fails to attack the allogeneic fetus. Two plasma proteins with described immunoregulatory activities, pregnancy zone protein (PZP) and placental protein-14 (PP14; also known as glycodelin-A), increase dramatically during pregnancy, prompting us to examine their potential role in mediating fetal protection. First, we demonstrated that both native PZP and its receptor-recognized monoamine-activated form (MA-PZP) bound non-covalently and specifically to PP14, exhibiting K(d) values greater than 3 microM, as determined by surface plasmon resonance. Our evidence further suggests that PZP is potentially a more effective carrier of PP14 than its relative alpha2-macroglobulin. Second, we found that T-cell activation, as measured by increased proliferation and IL-2 production, was inhibited by either PZP or PP14 in a dose-dependent manner. However, when PZP and PP14 were combined, they acted synergistically to inhibit T cell proliferation and IL-2 production. Interestingly, the combination of PZP and PP14 had little effect on the production of T(H)2 cytokine, IL-4. Based upon these findings, we hypothesize that PZP and PP14 form a stable complex in the plasma of pregnant women and together act synergistically to selectively modulate T-cell activation. Mechanistically, this activity appears to be independent of the PZP receptor (CD91) or PZP's anti-proteinase activity.


Subject(s)
Carrier Proteins/physiology , Cytokines/biosynthesis , Glycoproteins/physiology , Pregnancy Proteins/physiology , T-Lymphocytes/immunology , Antigens, CD/drug effects , Antigens, CD/immunology , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell Proliferation/drug effects , Cytokines/drug effects , Female , Glycodelin , Glycoproteins/immunology , Glycoproteins/metabolism , Humans , Interleukin-2/biosynthesis , Interleukin-4/biosynthesis , Kinetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Low Density Lipoprotein Receptor-Related Protein-1 , Lymphocyte Activation/immunology , Pregnancy , Pregnancy Proteins/immunology , Pregnancy Proteins/metabolism , Protein Binding , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Time Factors , alpha-Macroglobulins/immunology , alpha-Macroglobulins/metabolism
20.
Immunol Res ; 27(2-3): 565-74, 2003.
Article in English | MEDLINE | ID: mdl-12857999

ABSTRACT

Antigen-presenting cells (APC) can be refaced with "protein paints" that change the appearance of their T cell-oriented trans signal arrays. Our group has developed three categories of protein paints suitable for this kind of APC engineering: artificial glycosylphosphatidylinositol (GPI) proteins, palmitated-protein A:Fc*1 fusion protein conjugates, and trans signal converter proteins. Protein paints have been devised with either immune enhancement or suppression in mind. Costimulator * GPI and palmitated-protein A costimulator * Fcgamma1 conjugates can be used to augment the immune-activating potential of tumor cells. Alternatively, protein paints can be designed to transform APC into artificial veto cells, in essence creating Trojan horses capable of inhibiting pathogenic T cells. Trans signal converter proteins (TSCP) have been devised for this purpose. Our first paradigmatic inhibitory TSCP, CTLA-4 * Fas ligand, binds to APC, and in so doing, simultaneously blocks B7 costimulation (via CTLA-4) and sends inhibitory trans signals (via Fas ligand) to T cells with dramatic efficacy. Protein transfer offers a number of advantages over gene transfer in facilitating quantitative and combinatorial protein expression and simplifying in vivo applications; the palette of protein paints with immunotherapeutic potential will undoubtedly continue to evolve.


Subject(s)
Antigen-Presenting Cells/immunology , Cancer Vaccines/immunology , Membrane Proteins/immunology , Protein Engineering/methods , Animals , Humans , Interleukin-2/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...