Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Andrology ; 10(4): 789-799, 2022 05.
Article in English | MEDLINE | ID: mdl-35224888

ABSTRACT

BACKGROUND: Glucose-dependent insulinotropic polypeptide receptor (Gipr) gene expression has been reported in mouse spermatids and Gipr knockout male mice have previously been reported to have decreased in vitro fertilization, although the role of Gipr signaling in male mouse fertility is not well understood. OBJECTIVES: The purposes of these studies were to determine the role of glucose-dependent insulinotropic polypeptide receptor in male fertility using Gipr knockout mice and anti-glucose-dependent insulinotropic polypeptide receptor antibody-treated wild-type mice and to determine if the expression of Gipr in mouse testes is similar in non-human and human primates. METHODS AND MATERIALS: Adiponectin promoter-driven Gipr knockout male mice (GiprAdipo-/- ) were assessed for in vitro and in vivo fertility, sperm parameters, and testicular histology. CD1 male mice were administered an anti-glucose-dependent insulinotropic polypeptide receptor antibody (muGIPR-Ab) prior to and during mating for assessment of in vivo fertility and sperm parameters. Expression of Gipr/GIPR mRNA in the mouse, cynomolgus monkey, and human testes was assessed by in situ hybridization methods using species-specific probes. RESULTS: GiprAdipo-/- male mice are infertile in vitro and in vivo, despite normal testis morphology, sperm counts, and sperm motility. In contrast, administration of muGIPR-Ab to CD1 male mice did not impact fertility. While Gipr mRNA expression is detectable in the mouse testes, GIPR mRNA expression is not detectable in monkey or human testes. DISCUSSION: The infertility of GiprAdipo-/- male mice correlated with the lack of Gipr expression in the testis and/or adipocyte tissue. However, as administration of muGIPR-Ab did not impact the fertility of adult male mice, it is possible that the observations in genetically deficient male mice are related to Gipr deficiency during development. CONCLUSION: Our data support a role for Gipr expression in the mouse testis during the development of sperm fertilization potential, but based on gene expression data, a similar role for glucose-dependent insulinotropic polypeptide receptor in non-human primate or human male fertility is unlikely.


Subject(s)
Gastric Inhibitory Polypeptide , Testis , Animals , Female , Fertility , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/metabolism , Gene Expression , Humans , Macaca fascicularis/genetics , Macaca fascicularis/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Gastrointestinal Hormone , Sperm Motility , Testis/metabolism
2.
J Immunotoxicol ; 17(1): 110-121, 2020 12.
Article in English | MEDLINE | ID: mdl-32525431

ABSTRACT

Mast cells play key roles in allergy, anaphylaxis/anaphylactoid reactions, and defense against pathogens/toxins. These cells contain cytoplasmic granules with a wide spectrum of pleotropic mediators that are released upon activation. While mast cell degranulation (MCD) occurs upon clustering of the IgE receptor bound to IgE and antigen, MCD is also triggered through non-IgE-mediated mechanisms, one of which is via Mas-related G protein-coupled receptor X2 (MRGPRX2). MRGPRX2 can be activated by many basic biogenic amines and peptides. Consequently, MRGPRX2-mediated MCD is an important potential safety liability for peptide therapeutics. To facilitate peptide screening for this liability in early preclinical drug development, a rapid, high-throughput engineered CHO-K1 cell-based MRGPRX2 activation assay was evaluated and compared to histamine release in CD34+ stem cell-derived mature human mast cells as a reference assay, using 30 positive control and 29 negative control peptides for MCD. Both G protein-dependent (Ca2+ endpoint) and -independent (ß-arrestin endpoint) pathways were assessed in the MRGPRX2 activation assay. The MRGPRX2 activation assay had a sensitivity of 100% for both Ca2+ and ß-arrestin endpoints and a specificity of 93% (ß-arrestin endpoint) and 83% (Ca2+ endpoint) compared to histamine release in CD34+ stem cell-derived mature human mast cells. These findings suggest that assessing MRGPRX2 activation in an engineered cell model can provide value as a rapid, high-throughput, economical mechanism-based screening tool for early MCD hazard identification during preclinical safety evaluation of peptide-based therapeutics.


Subject(s)
Cell Degranulation/drug effects , High-Throughput Screening Assays/methods , Mast Cells/drug effects , Nerve Tissue Proteins/metabolism , Peptides/adverse effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Antigens, CD34/metabolism , Cell Degranulation/immunology , Cell Engineering , Cells, Cultured , Cytotoxicity Tests, Immunologic/methods , Drug Evaluation, Preclinical/methods , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Histamine/analysis , Histamine/metabolism , Humans , Mast Cells/immunology , Mast Cells/metabolism , Primary Cell Culture , Sensitivity and Specificity
3.
Cytokine X ; 2(4): 100042, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33458650

ABSTRACT

Immunomodulatory therapeutics such as monoclonal antibodies (mAb) carry an inherent risk of undesired immune reactions. One such risk is cytokine release syndrome (CRS), a rapid systemic inflammatory response characterized by the secretion of pro-inflammatory cytokines from immune cells. It is crucial for patient safety to correctly identify potential risk of CRS prior to first-in-human dose administration. For this purpose, a variety of in vitro cytokine release assays (CRA) are routinely used as part of the preclinical safety assessment of novel therapeutic mAbs. One of the challenges for the development and comparison of CRA performance is the lack of availability of standard positive and negative control mAbs for use in assay qualification. To address this issue, the National Institute for Biological Standards and Control (NIBSC) developed a reference panel of lyophilised mAbs known to induce CRS in the clinic: human anti-CD52, mouse anti-CD3 and human superagonistic (SA) anti-CD28 mAb manufactured according to the respective published sequences of Campath-1H® (alemtuzumab, IgG1) , Orthoclone OKT-3® (muromonab, IgG2a) and TGN1412 (theralizumab, IgG4), as well as three isotype matched negative controls (human IgG1, mouse IgG2a and human IgG4, respectively). The relative capacity of these control mAbs to stimulate the release of IFN-γ, IL-2, TNF-α and IL-6 in vitro was evaluated in eleven laboratories in an international collaborative study mediated through the HESI Immuno-safety Technical Committee Cytokine Release Assay Working Group. Participants tested the NIBSC mAbs in a variety of CRA platforms established at each institution. This paper presents the results from the centralised cytokine quantification on all the plasma/supernatants corresponding to the stimulation of immune cells in the different CRA platforms by a single concentration of each mAb. Each positive control mAb induced significant cytokine release in most of the tested CRA platforms. There was a high inter-laboratory variability in the levels of cytokines produced, but similar patterns of response were observed across laboratories that replicated the cytokine release patterns previously published for the respective clinical therapeutic mAbs. Therefore, the positive and negative mAbs are suitable as a reference panel for the qualification and validation of CRAs, comparison of different CRA platforms (e.g. solid vs aqueous phase), and intra- and inter-laboratory comparison of CRA performance. Thus, the use of this panel of positive and negative control mAbs will increase the confidence in the robustness of a CRA platform to identify a potential CRS risk for novel immunomodulatory therapeutic candidates.

4.
Endocr Rev ; 41(1)2020 01 01.
Article in English | MEDLINE | ID: mdl-31511854

ABSTRACT

Glucose-dependent insulinotropic polypeptide receptor (GIPR) is associated with obesity in human genome-wide association studies. Similarly, mouse genetic studies indicate that loss of function alleles and glucose-dependent insulinotropic polypeptide overexpression both protect from high-fat diet-induced weight gain. Together, these data provide compelling evidence to develop therapies targeting GIPR for the treatment of obesity. Further, both antagonists and agonists alone prevent weight gain, but result in remarkable weight loss when codosed or molecularly combined with glucagon-like peptide-1 analogs preclinically. Here, we review the current literature on GIPR, including biology, human and mouse genetics, and pharmacology of both agonists and antagonists, discussing the similarities and differences between the 2 approaches. Despite opposite approaches being investigated preclinically and clinically, there may be viability of both agonists and antagonists for the treatment of obesity, and we expect this area to continue to evolve with new clinical data and molecular and pharmacological analyses of GIPR function.


Subject(s)
Anti-Obesity Agents/therapeutic use , Molecular Targeted Therapy , Obesity/drug therapy , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Animals , Genome-Wide Association Study , Humans , Mice , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Obesity/genetics , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/physiology
5.
Toxicol Sci ; 170(1): 10-19, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31020324

ABSTRACT

Natural killer (NK) cells are lymphocytes capable of cytotoxicity against virally infected cells and tumor cells. The display of effector function by NK cells is the result of interactions between germline encoded activating/inhibitory NK cell receptors and their ligands (major histocompatibility complex class I, major histocompatibility complex class I-like, viral, and cellular stress-related surface molecules) expressed on target cells. Determination of NK cell number and function is a common element of the immunotoxicology assessment paradigm for the development of certain classes of pharmaceuticals across a range of modalities. This article summarizes the evidence associating NK cell dysfunction with infectious and cancer risks, reviews emerging NK cell biology, including the impact of immunogenetics on NK cell education and function, and provides perspectives about points to consider when assessing NK cell function in different species in the context of safety assessment.


Subject(s)
Disease Resistance/immunology , Drug-Related Side Effects and Adverse Reactions/immunology , HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/drug effects , Receptors, Natural Killer Cell/metabolism , Animals , Cytotoxicity, Immunologic , Disease Resistance/drug effects , Dose-Response Relationship, Drug , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Ligands , Pharmaceutical Preparations/administration & dosage
6.
J Biol Chem ; 292(5): 1876-1883, 2017 02 03.
Article in English | MEDLINE | ID: mdl-27994063

ABSTRACT

The stable effector functionLess (SEFL) antibody was designed as an IgG1 antibody with a constant region that lacks the ability to interact with Fcγ receptors. The engineering and stability and pharmacokinetic assessments of the SEFL scaffold is described in the accompanying article (Jacobsen, F. W., Stevenson, R., Li, C., Salimi-Moosavi, H., Liu, L., Wen, J., Luo, Q., Daris, K., Buck, L., Miller, S., Ho, S-Y., Wang, W., Chen, Q., Walker, K., Wypych, J., Narhi, L., and Gunasekaran, K. (2017) J. Biol. Chem 292). The biological properties of these SEFL antibodies were assessed in a variety of human and cynomolgus monkey in vitro assays. Binding of parent molecules and their SEFL variants to human and cynomolgus monkey FcγRs were evaluated using flow cytometry-based binding assays. The SEFL variants tested showed decreased binding affinity to human and cynomolgus FcγRs compared with the wild-type IgG1 antibody. In addition, SEFL variants demonstrated no antibody-dependent cell-mediated cytotoxicity in vitro against Daudi cells with cynomolgus monkey peripheral blood mononuclear cells, and had minimal complement-dependent cytotoxicity activity similar to that of the negative control IgG2 in a CD20+ human Raji lymphoma cell line. SEFL mutations eliminated off-target antibody-dependent monocyte phagocytosis of cynomolgus monkey platelets, and cynomolgus platelet activation in vitro These experiments demonstrate that the SEFL modifications successfully eliminated Fc-associated effector binding and functions.


Subject(s)
Antibodies, Monoclonal , Blood Platelets/immunology , Immunoglobulin G , Monocytes/immunology , Phagocytosis/drug effects , Platelet Activation/drug effects , Receptors, IgG , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Macaca fascicularis , Mice , Phagocytosis/immunology , Platelet Activation/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology
7.
Cytokine ; 85: 101-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27309676

ABSTRACT

In October 2013, the International Life Sciences Institute - Health and Environmental Sciences Institute Immunotoxicology Technical Committee (ILSI-HESI ITC) held a one-day workshop entitled, "Workshop on Cytokine Release: State-of-the-Science, Current Challenges and Future Directions". The workshop brought together scientists from pharmaceutical, academic, health authority, and contract research organizations to discuss novel approaches and current challenges for the use of in vitro cytokine release assays (CRAs) for the identification of cytokine release syndrome (CRS) potential of novel monoclonal antibody (mAb) therapeutics. Topics presented encompassed a regulatory perspective on cytokine release and assessment, case studies regarding the translatability of preclinical cytokine data to the clinic, and the latest state of the science of CRAs, including comparisons between mAb therapeutics within one platform and across several assay platforms, a novel physiological assay platform, and assay optimization approaches such as determination of FcR expression profiles and use of statistical tests. The data and approaches presented confirmed that multiple CRA platforms are in use for identification of CRS potential and that the choice of a particular CRA platform is highly dependent on the availability of resources for individual laboratories (e.g. positive and negative controls, number of human blood donors), the assay through-put required, and the mechanism-of-action of the therapeutic candidate to be tested. Workshop participants agreed that more data on the predictive performance of CRA platforms is needed, and current efforts to compare in vitro assay results with clinical cytokine assessments were discussed. In summary, many laboratories continue to focus research efforts on the improvement of the translatability of current CRA platforms as well explore novel approaches which may lead to more accurate, and potentially patient-specific, CRS prediction in the future.


Subject(s)
Cytokines/blood , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biological Assay/methods , Humans , Immune System Diseases/blood , Immune System Diseases/drug therapy
8.
Toxicol Pathol ; 42(1): 286-92, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24190913

ABSTRACT

To assess relative sensitivity for detection of cytokines and chemokines in cynomolgus serum samples, we tested three commercially available multiplex array kits using the Luminex® platform with sera from animals exposed by intravenous injection to 150 µg/kg staphylococcal enterotoxin B (SEB) or 20 µg/kg lipopolysaccharide (LPS). Each of these kits detected similar patterns of changes in circulating cytokines/chemokines in response to SEB or LPS stimulation, especially the induction of high amounts of interleukin (IL)-2 and interferon-gamma (IFN-γ) in response to SEB but not LPS. However, there were clear differences in sensitivity for particular analytes, especially for IL-10. Additional experiments that focused on one multiplex array kit demonstrated very low or undetectable levels of cytokines in naive cynomolgus macaques, except for highly variable background levels of IL-8, monocyte chemoattractant protein-1, and macrophage inflammatory protein-1ß. Therefore, multiplex array analysis of circulating cytokine/chemokine patterns was capable of detection of systemic activation of diverse immune cell subsets.


Subject(s)
Chemokine CCL2/blood , Interferon-gamma/blood , Interleukin-10/blood , Interleukin-2/blood , Interleukin-8/blood , Protein Array Analysis/methods , Administration, Intravenous , Animals , Chemokine CCL4/blood , Enterotoxins/administration & dosage , Enterotoxins/adverse effects , Female , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/adverse effects , Macaca fascicularis/immunology , Male , Reagent Kits, Diagnostic
9.
Toxicol Pathol ; 41(7): 951-69, 2013.
Article in English | MEDLINE | ID: mdl-23475561

ABSTRACT

Cynomolgus monkeys dosed with a therapeutic monoclonal antibody (mAbY.1) at ≥ 50 mg/kg had unexpected acute thrombocytopenia (nadir ~3,000 platelets/µl), sometimes with decreases in red cell mass. Increased activated macrophages, mitotic figures, and erythrophagocytosis were observed in the spleen. Binding of mAbY.1 to cynomolgus peripheral blood cells could not be detected in vitro. mAbY.1 induced phagocytosis of platelets by peripheral blood monocytes from cynomolgus monkeys, but not from humans. mAbs sharing the same constant domain (Fc) sequences, but differing from mAbY.1 in their variable domains, bound competitively to and had similar biological activity against the intended target. None of these antibodies had hematologic liabilities in vitro or in vivo. Neither the F(ab')2 portion of mAbY.1 nor the F(ab')2 portion on an aglycosylated Fc (IgG1) framework caused phagocytosis of platelets in vitro. These data suggest that the hematologic effects of mAbY.1 in cynomolgus monkeys likely occurred through an off-target mechanism, shown to be driven by 1 to 3 amino acid differences in the light chain. The hematologic effects made mAbY.1 an unsuitable candidate for further development as a therapeutic agent. This example demonstrates that nonclinical safety studies may be essential for understanding off-target effects of mAbs prior to clinical trials.


Subject(s)
Anemia/chemically induced , Antibodies, Monoclonal/toxicity , Thrombocytopenia/chemically induced , Anemia/blood , Animals , Antibodies, Monoclonal/administration & dosage , Blood Platelets/pathology , Female , Humans , Macaca fascicularis , Macrophage Activation , Male , Phagocytosis , Reticulocytes/pathology , Spleen/drug effects , Spleen/pathology , Thrombocytopenia/blood
10.
Toxicol Pathol ; 40(6): 899-917, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22552394

ABSTRACT

AMG X, a human neutralizing monoclonal antibody (mAb) against a soluble human protein, caused thrombocytopenia, platelet activation, reduced mean arterial pressure, and transient loss of consciousness in cynomolgus monkeys after first intravenous administration. In vitro, AMG X induced activation in platelets from macaque species but not from humans or baboons. Other similar mAbs against the same pharmacological target failed to induce these in vivo and in vitro effects. In addition, the target protein was known to not be expressed on platelets, suggesting that platelet activation occurred through an off-target mechanism. AMG X bound directly to cynomolgus platelets and required both the Fab and Fc portion of the mAb for platelet activation. Binding to platelets was inhibited by preincubation of AMG X with its pharmacological target or with anti-human Fc antibodies or by preincubation of platelets with AMG X F(ab')(2) or human immunoglobulin (IVIG). AMG X F(ab')(2) did not activate platelets. Thus, platelet activation required both recognition/binding of a platelet ligand with the Fab domain and interaction of platelet Fc receptors (i.e., FcγRIIa) with the Fc domain. These findings reflect the complexity of the mechanism of action of mAbs and the increasing awareness of potential for unintended effects in preclinical species.


Subject(s)
Antibodies, Monoclonal/toxicity , Blood Platelets/drug effects , Platelet Activation/drug effects , Administration, Intravenous , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Blood Platelets/metabolism , Humans , Hypotension/blood , Hypotension/chemically induced , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fc Fragments/metabolism , Macaca fascicularis , Male , Papio , Platelet Aggregation/drug effects , Protein Binding , Serotonin/metabolism , Syncope/blood , Syncope/chemically induced , Thrombocytopenia/blood , Thrombocytopenia/chemically induced , Thromboxane B2/metabolism
11.
Toxicol Pathol ; 40(1): 107-12, 2012.
Article in English | MEDLINE | ID: mdl-22033502

ABSTRACT

Regulatory T cells (Tregs) are a rare subset of lymphocytes that inhibit the activation and effector functions of T cells and are important regulators of immune responses. Although Tregs are well characterized in humans and rodents, little is known about their immunophenotyping (IP) profile in cynomolgus macaques (Macaca fascicularis), which is an important species for pharmacological and toxicological evaluation of potential immune modulators because of their similar physiologic, genetic, and metabolic response patterns to humans. The authors have developed an immunophenotyping panel using a high-throughput 96-well microtiter plate-based assay to detect circulating Tregs (CD3(+)CD4(+)CD25(hi)FoxP3(+)) and have determined the normal range for the number of Tregs in naive healthy cynomolgus macaques to be 56.4 to 179.7 cells/µL (mean ± SEM = 113.6 ± 5.1 cells/µL; n = 25). Furthermore, the authors compared the resulting FoxP3(+) Treg profiles with a CD127(lo) cell-surface panel (CD3(+)CD4(+)CD25(hi) CD127(lo)) and found a close correlation between the absolute numbers of CD3(+)CD4(+)CD25(hi)FoxP3(+) and CD3(+)CD4(+)CD25(hi)CD127(lo) cells (mean ± SD = 120 ± 8.0 cells/µL). Quantification of circulating Tregs in cynomolgus macaques in this high-throughput assay may help to identify drug candidates that affect this rare, but critical, immunoregulatory cell population.


Subject(s)
Flow Cytometry/methods , Immunophenotyping/methods , Macaca fascicularis/blood , T-Lymphocytes, Regulatory/cytology , Animals , Antigens, CD/blood , Female , Forkhead Transcription Factors/blood , High-Throughput Screening Assays , Macaca fascicularis/immunology , Male , Models, Animal , T-Lymphocytes, Regulatory/chemistry
12.
J Immunotoxicol ; 9(1): 34-42, 2012.
Article in English | MEDLINE | ID: mdl-22074378

ABSTRACT

Human peripheral blood mononuclear cells (PBMC) are routinely used in vitro to detect cytokine secretion as part of preclinical screens to delineate agonistic and antagonistic action of therapeutic monoclonal antibodies (mAbs). Preclinical value of standard human PBMC assays to detect cytokine release syndrome (CRS) has been questioned, as they did not predict the "cytokine storm" that occurred when healthy human volunteers were given a CD28-specific super-agonist mAb, TGN1412. In this article, we describe a three-dimensional biomimetic vascular test-bed that can be used as a more physiologically relevant assay for testing therapeutic Abs. For developing such a system, we used TGN1412 as a model mAb. We tested soluble TGN1412 on various combinations of human blood components in a module containing endothelial cells grown on a collagen scaffold and measured cytokine release using multiplex array. Our system, consisting of whole leukocytes, endothelial cells, and 100% autologous platelet-poor plasma (PPP) consistently produced proinflammatory cytokines in response to soluble TGN1412. In addition, other mAb therapeutics known to induce CRS or first infusion reactions, such as OKT3, Campath-1H, or Herceptin, generated cytokine profiles in our model system consistent with their in vivo responses. As a negative control we tested the non-CRS mAbs Avastin and Remicade and found little difference between these mAbs and the placebo control. Our data indicate that this novel assay may have preclinical value for predicting the potential of CRS for mAb therapeutics.


Subject(s)
Antibodies, Monoclonal, Humanized/toxicity , Biomimetics , Cytokines/metabolism , Drug Evaluation, Preclinical/methods , Granulocytes/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Leukocytes, Mononuclear/drug effects , Toxicity Tests , Cells, Cultured , Coculture Techniques , Collagen , Granulocytes/immunology , Granulocytes/metabolism , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Plasma , Risk Assessment , Time Factors , Tissue Scaffolds
13.
Toxicol Sci ; 117(2): 253-62, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20484381

ABSTRACT

Regulatory T cells (Tregs) constitute a subset of lymphocytes that have the capability of suppressing immune responses in vivo and in vitro both directly by cell-cell contact and indirectly through the production of anti-inflammatory cytokines, such as interleukin-10 and tumor growth factor-ß. Tregs constitute a small subset of T lymphocytes, yet their presence can prevent and control autoimmune disease and organ transplant rejection and contribute to maternal tolerance of fetal alloantigens, whereas their absence results in uncontrolled inflammation. But Treg function may not always be considered beneficial: There is growing evidence that the immunosuppressive effects of Tregs are also associated with growth of tumor cells. Thus, Tregs are of considerable medical interest as targets for the treatment of both inflammatory diseases and cancer. In this review of published literature, we describe some well-characterized immunomodulatory drugs and environmental toxicants that can either positively or negatively affect the number and/or function of Tregs in animal models and/or human patients. The targeted suppression or enhancement of Treg function needs to be carefully considered in immunotoxicity evaluations as manipulation of this immune cell population could result in undesired consequences, including decreased host resistance, decreased fertility, or increased incidence of inflammatory disease.


Subject(s)
Immune System/drug effects , Immunologic Factors/toxicity , T-Lymphocytes, Regulatory/drug effects , Animals , Cell Count , Disease Models, Animal , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Immune System/immunology , Mice , Species Specificity , T-Lymphocytes, Regulatory/immunology
14.
J Immunol ; 174(10): 6416-23, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15879143

ABSTRACT

Current evidence indicates that the chronic inflammation observed in the intestines of patients with inflammatory bowel disease is due to an aberrant immune response to enteric flora. We have developed a lipid A-mimetic, CRX-526, which has antagonistic activity for TLR4 and can block the interaction of LPS with the immune system. CRX-526 can prevent the expression of proinflammatory genes stimulated by LPS in vitro. This antagonist activity of CRX-526 is directly related to its structure, particularly secondary fatty acyl chain length. In vivo, CRX-526 treatment blocks the ability of LPS to induce TNF-alpha release. Importantly, treatment with CRX-526 inhibits the development of moderate-to-severe disease in two mouse models of colonic inflammation: the dextran sodium sulfate model and multidrug resistance gene 1a-deficient mice. By blocking the interaction between enteric bacteria and the innate immune system, CRX-526 may be an effective therapeutic molecule for inflammatory bowel disease.


Subject(s)
Adjuvants, Immunologic/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Glucosamine/analogs & derivatives , Glucosamine/pharmacology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/prevention & control , Lipid A/analogs & derivatives , Lipid A/pharmacology , Receptors, Immunologic/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adjuvants, Immunologic/chemical synthesis , Animals , Caproates/chemistry , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colitis/prevention & control , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Glucosamine/chemistry , HeLa Cells , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Receptors, Immunologic/metabolism , Toll-Like Receptor 4 , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
15.
J Clin Invest ; 113(9): 1296-306, 2004 May.
Article in English | MEDLINE | ID: mdl-15124021

ABSTRACT

Chronic intestinal inflammation, as seen in inflammatory bowel disease (IBD), results from an aberrant and poorly understood mucosal immune response to the microbiota of the gastrointestinal tract in genetically susceptible individuals. Here we used serological expression cloning to identify commensal bacterial proteins that could contribute to the pathogenesis of IBD. The dominant antigens identified were flagellins, molecules known to activate innate immunity via Toll-like receptor 5 (TLR5), and critical targets of the acquired immune system in host defense. Multiple strains of colitic mice had elevated serum anti-flagellin IgG2a responses and Th1 T cell responses to flagellin. In addition, flagellin-specific CD4(+) T cells induced severe colitis when adoptively transferred into naive SCID mice. Serum IgG to these flagellins, but not to the dissimilar Salmonella muenchen flagellin, was elevated in patients with Crohn disease, but not in patients with ulcerative colitis or in controls. These results identify flagellins as a class of immunodominant antigens that stimulate pathogenic intestinal immune reactions in genetically diverse hosts and suggest new avenues for the diagnosis and antigen-directed therapy of patients with IBD.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Crohn Disease/immunology , Flagellin/immunology , Inflammatory Bowel Diseases/etiology , Animals , Antigen-Presenting Cells/immunology , Bacterial Proteins/genetics , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Cecum/microbiology , Cells, Cultured , Cloning, Molecular , Crohn Disease/pathology , Dose-Response Relationship, Immunologic , Escherichia coli/metabolism , Flagellin/genetics , Humans , Immunoglobulin G/blood , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Lymphocyte Activation , Mice , Mice, Inbred C3H , Mice, Inbred CBA , Mice, SCID , Molecular Sequence Data , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...