Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
Immunohorizons ; 5(7): 581-589, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326199

ABSTRACT

The expression of Bruton tyrosine kinase (BTK) in B cells and innate immune cells provides essential downstream signaling for BCR, Fc receptors, and other innate immune cell pathways. The topical covalent BTK inhibitor PRN473 has shown durable, reversible BTK occupancy with rapid on-rate and slow off-rate binding kinetics and long residence time, resulting in prolonged, localized efficacy with low systemic exposure in vivo. Mechanisms of PRN473 include inhibition of IgE (FcεR)-mediated activation of mast cells and basophils, IgG (FcγR)-mediated activation of monocytes, and neutrophil migration. In vivo, oral PRN473 was efficacious and well tolerated in the treatment of canine pemphigus foliaceus. In this study, we evaluated in vitro selectivity and functionality, in vivo skin Ab inflammatory responses, and systemic pharmacology with topically administered PRN473. Significant dose-dependent inhibition of IgG-mediated passive Arthus reaction in rats was observed with topical PRN473 and was maintained when given 16 h prior to challenge, reinforcing extended activity with once-daily administration. Similarly, topical PRN473 resulted in significant dose-dependent inhibition of the mouse passive cutaneous anaphylaxis IgE-mediated reaction. Multiday treatment with topical PRN473 in rodents resulted in low-to-no systemic accumulation, suggesting that efficacy was mainly due to localized exposure. Reduced skin Ab inflammatory activity was also confirmed with oral PRN473. These preclinical studies provide a strong biologic basis for targeting innate immune cell responses locally in the skin, with rapid onset of action following once-daily topical PRN473 administration and minimal systemic exposure. Dose-dependent inhibition in these preclinical models of immune-mediated skin diseases support future clinical studies.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Arthus Reaction , Passive Cutaneous Anaphylaxis , Protein Kinase Inhibitors , Skin Diseases , Animals , Female , Humans , Mice , Rats , Administration, Cutaneous , Administration, Oral , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Arthus Reaction/drug therapy , Arthus Reaction/immunology , Arthus Reaction/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Passive Cutaneous Anaphylaxis/drug effects , Protein Kinase Inhibitors/administration & dosage , Skin/drug effects , Skin/immunology , Skin/pathology , Skin Diseases/drug therapy , Skin Diseases/immunology , Skin Diseases/pathology
2.
J Dermatol Sci ; 95(1): 36-43, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31262443

ABSTRACT

BACKGROUND: Local type III hypersensitivity reactions are acute inflammatory events induced by immune complex (IC) deposition. CD22 and CD72 are B cell-specific cell surface molecules that negatively regulate B cell function. OBJECTIVE: To elucidate the roles of CD22 and CD72 in the development of IgG-mediated type III hypersensitivity reactions. METHOD: The reverse Arthus reaction model in the skin was induced in mice lacking CD22 (CD22-/-), CD72 (CD72-/-), and both of them (CD22-/-/CD72-/-). Edema at 4h and hemorrhage at 8h after IC challenge were evaluated. Inflammatory cell infiltration and cytokine and chemokine expression were also examined. RESULTS: Edema and hemorrhage were significantly reduced in CD22-/-/CD72-/- mice compared with wild-type mice. The loss of both membrane proteins resulted in a greater decrease in edema at 4h, but not hemorrhage at 8h, than the loss of each protein alone. Infiltration of neutrophils, macrophages, and T cells, and the expression of TNF-α, IL-6, MIP-1α, and CCR5 mRNA were also diminished in the knockout mice compared to wild-type mice, and most significantly reduced in CD22-/-/CD72-/- mice. Regulatory T (Treg) cells in the spleen were significantly increased in all knockout mice at 4h. Significant differences in the severity of edema and hemorrhage between wild-type and knockout mice were lost when Treg cells were depleted in the knockout mice. CONCLUSION: These results demonstrate that CD22 and CD72 expression contribute to the development of the reverse Arthus reaction model and CD22 and CD72 might be therapeutic targets for human IC-mediated diseases.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Arthus Reaction/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Skin/immunology , Animals , Antigen-Antibody Complex/administration & dosage , Antigen-Antibody Complex/immunology , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Arthus Reaction/pathology , Biopsy , Injections, Intradermal , Mice , Mice, Knockout , Sialic Acid Binding Ig-like Lectin 2/genetics , Skin/pathology
3.
Clin Exp Dermatol ; 41(8): 871-877, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27753135

ABSTRACT

BACKGROUND: Tumour necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a pro-inflammatory cytokine, which is closely associated with the pathogenesis of various types of cutaneous vasculitis (CV). AIM: To investigate the therapeutic effects of an anti-TWEAK monoclonal antibody (mAb) in a mouse model of cutaneous reverse passive Arthus (RPA) reaction. METHODS: Cutaneous RPA reaction was induced in BALB/c mice by intradermal injection of anti-ovalbumin IgG into the left ear followed immediately by intravenous injection of chicken ovalbumin. After treatment, haemorrhagic lesions in the mouse skin were scored semiquantitatively. The amount of extravasated fluorescein isothiocyanate (FITC)-labelled bovine serum albumin (BSA) in the ears was detected spectrophotometrically. Expression of myeloperoxidase (MPO) was detected by immunohistochemical staining, while mRNA expression of TNF-α and interleukin (IL)-6 in lesional skin was detected by real-time quantitative (q)PCR. RESULTS: Our results indicated that anti-TWEAK mAb significantly attenuated the clinical and histopathological changes in immune complex (IC)-induced mice, and also reduced the semiquantitative haemorrhage score, FITC-labelled BSA extravasation and MPO activity. Real-time qPCR showed that anti-TWEAK mAb significantly inhibited mRNA expression of TNF-α and IL-6 in lesional skin from IC-induced mice. CONCLUSION: These data suggest that anti-TWEAK mAb can block vascular damage and leucocyte infiltration in IC-induced mice. TWEAK might be a candidate immunotherapeutic medicine for suppression of IC-induced CV.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthus Reaction/drug therapy , Cytokine TWEAK/antagonists & inhibitors , Skin Diseases/drug therapy , Animals , Arthus Reaction/metabolism , Arthus Reaction/pathology , Cytokine TWEAK/blood , Cytokines/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C , Peroxidase/metabolism , Real-Time Polymerase Chain Reaction , Skin Diseases/metabolism , Skin Diseases/pathology
4.
Am J Pathol ; 182(5): 1640-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23470165

ABSTRACT

A type III hypersensitivity reaction induced by an immune complex, such as leukocytoclastic vasculitis, is mediated by inflammatory cell infiltration that is highly regulated by multiple adhesion molecules. CX3CL1, a ligand for CX3C chemokine receptor 1 (CX3CR1), has recently been identified as a key mediator of leukocyte adhesion that functions without the recruitment of integrins or selectin-mediated rolling. To elucidate the role of CX3CL1 and CX3CR1 in the development of leukocytoclastic vasculitis, the cutaneous and peritoneal reverse Arthus reactions, classic experimental models for immune complex-mediated tissue injury, were examined in mice lacking CX3CR1. CX3CL1 expression in sera and lesional skin of patients with polyarteritis nodosa (PN) and healthy controls was also examined. Edema and hemorrhage were significantly reduced in CX3CR1(-/-) mice compared with wild-type mice. Infiltration of neutrophils and mast cells and expression of IL-6 and tumor necrosis factor-α were also decreased in CX3CR1(-/-) mice. CX3CL1 was expressed in endothelial cells during the cutaneous reverse Arthus reactions. Furthermore, serum CX3CL1 levels were significantly higher in patients with PN than in healthy controls. Endothelial cells in lesional skin of patients with PN strongly expressed CX3CL1. These results suggest that interactions between CX3CL1 and CX3CR1 may contribute to the development of leukocytoclastic vasculitis by regulating neutrophil and mast cell recruitment and cytokine expression.


Subject(s)
Antigen-Antibody Complex/metabolism , Chemokine CX3CL1/metabolism , Receptors, Chemokine/metabolism , Vasculitis/immunology , Vasculitis/pathology , Adult , Animals , Arthus Reaction/genetics , Arthus Reaction/immunology , Arthus Reaction/pathology , CX3C Chemokine Receptor 1 , Chemokine CX3CL1/blood , Edema/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation , Hemorrhage/pathology , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Middle Aged , Neutrophil Infiltration , Peritoneum/pathology , Polyarteritis Nodosa/blood , Polyarteritis Nodosa/immunology , Polyarteritis Nodosa/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Chemokine/deficiency , Skin/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vasculitis/blood , Vasculitis/genetics
5.
J Leukoc Biol ; 93(4): 573-84, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23381473

ABSTRACT

H2S has been highlighted recently as an endogenous, gaseous signaling molecule, especially in inflammations. The deposition of IC induces an acute inflammatory response with tissue injury. To assess the roles of H2S in the IC-induced diseases, the cutaneous, reverse passive Arthus reaction was conducted using NaHS as a H2S donor. Furthermore, we conducted similar experiments using selectin(-/-) mice to determine the involvement of selectin molecules in the H2S-mediated pathway. Exogenous application of NaHS dramatically attenuated inflammatory reactions in WT mice associated with Arthus reaction. Namely, mRNA expressions of TNF-α, IFN-γ, and neutrophil numbers were reduced significantly in the lesional skins of NaHS-treated WT mice relative to untreated ones. NaHS treatment significantly reduced these three parameters in the lesional skins of E- and P-selectin(-/-) mice but not in those of L-selectin(-/-) mice. Quite similar results were obtained in the blocking study using WT mice injected with mAb to E-, P-, and L-selectin. Our results indicated that the exogenous application of NaHS attenuates inflammatory responses in reverse passive Arthus reaction through a L-selectin-involved pathway but not through E- or P-selectin pathways.


Subject(s)
Arthus Reaction/prevention & control , Hydrogen Sulfide/pharmacology , L-Selectin/immunology , RNA, Messenger/blood , Skin/drug effects , Sulfides/pharmacology , Animals , Antibodies/pharmacology , Antigen-Antibody Complex/immunology , Arthus Reaction/genetics , Arthus Reaction/immunology , Arthus Reaction/pathology , E-Selectin/genetics , E-Selectin/immunology , Gene Deletion , Gene Expression , Hydrogen Sulfide/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , L-Selectin/genetics , Male , Mice , Mice, Knockout , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , P-Selectin/genetics , P-Selectin/immunology , RNA, Messenger/genetics , Signal Transduction/drug effects , Skin/immunology , Skin/pathology , Sulfides/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
6.
J Anesth ; 27(2): 261-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23096126

ABSTRACT

PURPOSE: Neutrophil recruitment to the inflammatory sites is regulated by a variety of adhesion molecules including ß2 integrins. The dependency of neutrophil recruitment on ß2 integrins is variable in different tissues, but has not yet been verified in the cutaneous passive reverse Arthus reaction. We examined this question and also evaluated the impact of isoflurane on neutrophil recruitment to the skin because we previously showed in vitro that isoflurane binds and inhibits ß2 integrins. METHODS: The dependency on ß2 integrins in neutrophil recruitment to the skin in the Arthus reaction was examined using αL, αM and ß2 knockout mice. Then, we evaluated the effect of isoflurane on neutrophil recruitment to the skin. In addition, the effects of isoflurane on neutrophil binding to intercellular adhesion molecule-1 (ICAM-1), one of the ß2 integrin ligands, were studied in vitro using cell adhesion assays. RESULTS: Neutrophil recruitment to the skin in the Arthus reaction model was totally dependent on ß2 integrins, as ß2 knockout mice completely abolished it. However, the defect of only one of the ß2 integrins was not sufficient to abolish neutrophil recruitment. Isoflurane reduced neutrophil recruitment to the skin by approximately 90 %. Also, isoflurane inhibited neutrophil adhesion to ß2 integrin ligand ICAM-1. CONCLUSIONS: We demonstrated that (1) neutrophil recruitment to the skin was totally dependent on ß2 integrins, and (2) isoflurane significantly impaired neutrophil recruitment. Based on the previous studies on the contribution of other adhesion molecules in neutrophil recruitment, it is likely that isoflurane at least partially affects on ß2 integrins in this model.


Subject(s)
Anesthetics, Inhalation/pharmacology , Arthus Reaction/prevention & control , Isoflurane/pharmacology , Neutrophil Infiltration/drug effects , Animals , Arthus Reaction/pathology , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cell Adhesion/drug effects , Flow Cytometry , In Vitro Techniques , Integrin beta Chains/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/metabolism , Skin/pathology
7.
J Immunol ; 190(1): 324-33, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23225882

ABSTRACT

Heterotrimeric G proteins of the Gα(i) family have been implicated in signaling pathways regulating cell migration in immune diseases. The Gα(i)-protein-coupled C5a receptor is a critical regulator of IgG FcR function in experimental models of immune complex (IC)-induced inflammation. By using mice deficient for Gα(i2) or Gα(i3), we show that Gα(i2) is necessary for neutrophil influx in skin and lung Arthus reactions and agonist-induced neutrophilia in the peritoneum, whereas Gα(i3) plays a less critical but variable role. Detailed analyses of the pulmonary IC-induced inflammatory response revealed several shared functions of Gα(i2) and Gα(i3), including mediating C5a anaphylatoxin receptor-induced activation of macrophages, involvement in alveolar production of chemokines, transition of neutrophils from bone marrow into blood, and modulation of CD11b and CD62L expression that account for neutrophil adhesion to endothelial cells. Interestingly, C5a-stimulated endothelial polymorphonuclear neutrophil transmigration, but not chemotaxis, is enhanced versus reduced in the absence of neutrophil Gα(i3) or Gα(i2), respectively, and knockdown of endothelial Gα(i2) caused decreased transmigration of wild-type neutrophils. These data demonstrate that Gα(i2) and Gα(i3) contribute to inflammation by redundant, overlapping, and Gα(i)-isoform-specific mechanisms, with Gα(i2) exhibiting unique functions in both neutrophils and endothelial cells that appear essential for polymorphonuclear neutrophil recruitment in IC disease.


Subject(s)
Acute Lung Injury/immunology , GTP-Binding Protein alpha Subunit, Gi2/physiology , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Arthus Reaction/genetics , Arthus Reaction/immunology , Arthus Reaction/pathology , Cell Adhesion/genetics , Cell Adhesion/immunology , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Down-Regulation/genetics , Down-Regulation/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , GTP-Binding Protein alpha Subunit, Gi2/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Neutrophils/immunology , Neutrophils/pathology , Up-Regulation/genetics , Up-Regulation/immunology
8.
Am J Pathol ; 176(1): 259-69, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20008131

ABSTRACT

Platelets have been shown to be important in inflammation, but their role in the cutaneous Arthus reaction remains unclear. To assess the role of platelets in this pathogenetic process, the cutaneous Arthus reaction was examined in wild-type mice and mice lacking E-selectin, P-selectin, or P-selectin glycoprotein ligand-1 (PSGL-1) with or without platelet depletion by busulfan, a bone marrow precursor cell-specific toxin. Edema and hemorrhage induced by immune complex challenge significantly decreased in busulfan-treated wild-type mice compared with untreated mice. Busulfan treatment did not affect edema and hemorrhage in P-selectin- or PSGL-1-deficient mice, suggesting that the effect by busulfan is dependent on P-selectin and PSGL-1 expression. The inhibited edema and hemorrhage paralleled reduced infiltration of neutrophils and mast cells and reduced levels of circulating platelets. Increased cutaneous production of interleukin-6, tumor necrosis factor-alpha, and platelet-derived chemokines during Arthus reaction was inhibited in busulfan-treated wild-type mice relative to untreated mice, which paralleled the reduction in cutaneous inflammation. Flow cytometric analysis showed that immune complex challenge generated blood platelet-leukocyte aggregates that decreased by busulfan treatment. In thrombocytopenic mice, the cutaneous inflammation after immune complex challenge was restored by platelet infusion. These results suggest that platelets induce leukocyte recruitment into skin by forming platelet-leukocyte aggregates and secreting chemokines at inflamed sites, mainly through the interaction of P-selectin on platelets with PSGL-1 on leukocytes.


Subject(s)
Arthus Reaction/pathology , Blood Platelets/pathology , Cell Movement , Leukocytes/pathology , Skin/pathology , Animals , Blood Platelets/drug effects , Busulfan/administration & dosage , Busulfan/pharmacology , Cell Aggregation/drug effects , Cell Count , Cell Movement/drug effects , Chemokines/genetics , Chemokines/metabolism , Disease Models, Animal , Edema/pathology , Flow Cytometry , Gene Expression Regulation/drug effects , Hemorrhage/pathology , Inflammation Mediators/metabolism , Leukocytes/drug effects , Mast Cells/drug effects , Mast Cells/pathology , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
J Clin Invest ; 118(7): 2438-47, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18568075

ABSTRACT

Neutrophil granulocytes form the body's first line of antibacterial defense, but they also contribute to tissue injury and noninfectious, chronic inflammation. Proteinase 3 (PR3) and neutrophil elastase (NE) are 2 abundant neutrophil serine proteases implicated in antimicrobial defense with overlapping and potentially redundant substrate specificity. Here, we unraveled a cooperative role for PR3 and NE in neutrophil activation and noninfectious inflammation in vivo, which we believe to be novel. Mice lacking both PR3 and NE demonstrated strongly diminished immune complex-mediated (IC-mediated) neutrophil infiltration in vivo as well as reduced activation of isolated neutrophils by ICs in vitro. In contrast, in mice lacking just NE, neutrophil recruitment to ICs was only marginally impaired. The defects in mice lacking both PR3 and NE were directly linked to the accumulation of antiinflammatory progranulin (PGRN). Both PR3 and NE cleaved PGRN in vitro and during neutrophil activation and inflammation in vivo. Local administration of recombinant PGRN potently inhibited neutrophilic inflammation in vivo, demonstrating that PGRN represents a crucial inflammation-suppressing mediator. We conclude that PR3 and NE enhance neutrophil-dependent inflammation by eliminating the local antiinflammatory activity of PGRN. Our results support the use of serine protease inhibitors as antiinflammatory agents.


Subject(s)
Inflammation/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Leukocyte Elastase/metabolism , Myeloblastin/metabolism , Animals , Antigen-Antibody Complex/pharmacology , Arthus Reaction/metabolism , Arthus Reaction/pathology , Arthus Reaction/prevention & control , Cell Movement/drug effects , Chemotaxis, Leukocyte/drug effects , Granulins , Inflammation/drug therapy , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Leukocyte Elastase/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , Models, Biological , Myeloblastin/genetics , Neutrophil Activation/drug effects , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/metabolism , Ovalbumin/immunology , Progranulins , Respiratory Burst/drug effects , Tetradecanoylphorbol Acetate/pharmacology
10.
J Immunol ; 180(1): 580-9, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18097060

ABSTRACT

Binding of Ag-Ab immune complexes to cellular FcgammaR promotes cell activation, release of inflammatory mediators, and tissue destruction characteristic of autoimmune disease. To evaluate whether a soluble FcgammaR could block the proinflammatory effects of immune complexes, recombinant human (rh) versions of FcgammaRIA, FcgammaRIIA, and FcgammaRIIIA were prepared. Binding of rh-FcgammaRIA to IgG was of high affinity (KD=1.7x10(-10) M), whereas rh-FcgammaRIIA and rh-FcgammaRIIIA bound with low affinity (KD=0.6-1.9x10(-6) M). All rh-FcgammaR reduced immune complex precipitation, blocked complement-mediated lysis of Ab-sensitized RBC, and inhibited immune complex-mediated production of IL-6, IL-13, MCP-1, and TNF-alpha by cultured mast cells. Local or systemic delivery only of rh-FcgammaRIA, however, reduced edema and neutrophil infiltration in the cutaneous Arthus reaction in mice. 125I-labeled rh-FcgammaRIA was cleared from mouse blood with a rapid distribution phase followed by a slow elimination phase with a t1/2gamma of approximately 130 h. The highest percentage of injected radioactivity accumulated in blood approximately liver approximately carcass>kidney. s.c. dosing of rh-FcgammaRIA resulted in lower serum levels of inflammatory cytokines and prevented paw swelling and joint damage in a murine model of collagen Ab-induced arthritis. These data demonstrate that rh-FcgammaRIA is an effective inhibitor of type III hypersensitivity.


Subject(s)
Immune Complex Diseases/drug therapy , Receptors, IgG/therapeutic use , Animals , Antigen-Antibody Complex/drug effects , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthus Reaction/drug therapy , Arthus Reaction/pathology , Complement System Proteins/immunology , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Humans , Immune Complex Diseases/pathology , Immunoglobulin G/metabolism , Mast Cells/immunology , Mice , Receptors, IgG/biosynthesis
11.
Ann N Y Acad Sci ; 1109: 66-83, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17785292

ABSTRACT

Antigen-antibody (Ab) interactions that lead to the formation of immune complexes (ICs) are subtle biochemical processes determining health or disease according to the outcome. Good laboratory practice (GLP)-acknowledged IC detection methods reveal that plasma levels of up to 15 microg/mL heat-aggregated immunoglobulin G (IgG) equivalents are normal, indicating the physiological role of ICs. Among the major variables that influence the equilibrium association constant Ka, are specificity and epitope density of the antigen, Ig class/subclass of the Ab, IC complement (C)-activating capacity, Fc receptor (FcR) interaction, and cytokine activation pattern. The Ka of antigen-Ab binding at approximately 20 degrees C ranges from low affinity (105) to high affinity (107-1011). Beneficial ICs serve to remove and/or neutralize infectious or toxic antigens, following an infectious attack in immune and vaccinated hosts. Circulating ICs are more prone for benefit than tissue-bound ICs, which reflect in situ formation and/or undesired deposition in tissues due to overflow from insufficient reticuloendothelial system (RES) removal. The classical textbook topic on ICs still holds true but is under revision because of the improved knowledge of effector systems, such as C, cytokine, and FcR apparatus. Therapeutic options to treat IC-associated diseases include intravenous immunoglobulins (IVIG) at their onset and monoclonal antibodies (mAb) directed at C activation products and/or cytokines.


Subject(s)
Antigen-Antibody Complex/immunology , Immune System Diseases/immunology , Immune System Diseases/physiopathology , Animals , Antigen-Antibody Complex/blood , Arthus Reaction/blood , Arthus Reaction/immunology , Arthus Reaction/pathology , Chemical Phenomena , Chemistry, Physical , Cytokines/immunology , Humans , Protein Engineering
12.
Ann N Y Acad Sci ; 1109: 106-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17785295

ABSTRACT

We report an unusual case that highlights the clinical problems associated with autoimmune phenomena. A female (born 1972) was referred to our hospital with systemic lupus erythematosus (SLE) diagnosis. During the follow-up (7 years), we observed the appearance and the disappearance of a lot of autoantibodies detected. The history of recurrent bacterial sinopulmonary infections since puberty and enlargement of lymphonodes, elevated IgM, very low IgA and normal IgG levels, and the variable autoantibody profile oriented toward a "defective Ig class switch recombination" disorder: the hyper-IgM syndrome. Immunodeficiency and autoimmune phenomena may occur concomitantly in the same individual and sometimes the differential diagnosis is difficult.


Subject(s)
Autoimmunity/immunology , Hyper-IgM Immunodeficiency Syndrome/immunology , Antibodies/immunology , Antigens/immunology , Arthus Reaction/blood , Arthus Reaction/immunology , Arthus Reaction/pathology , Female , Humans , Hyper-IgM Immunodeficiency Syndrome/blood , Hyper-IgM Immunodeficiency Syndrome/diagnosis
13.
J Leukoc Biol ; 81(5): 1197-204, 2007 May.
Article in English | MEDLINE | ID: mdl-17299025

ABSTRACT

Immune complex (IC)-induced inflammation is mediated by inflammatory cell infiltration, a process that is highly regulated by expression of multiple adhesion molecules. The roles and interactions of ICAM-1 and VCAM-1, the major regulators of leukocyte firm adhesion, were examined in the cutaneous reverse-passive Arthus reaction using ICAM-1-deficient (ICAM-1-/-) mice and blocking mAb against VCAM-1. Within 8 h, IC challenge of wild-type mice induced edema, hemorrhage, interstitial accumulation of neutrophils and mast cells, as well as production of TNF-alpha and IL-6. All of these inflammatory parameters were reduced significantly in ICAM-1-/- mice. The blockade of VCAM-1 in wild-type mice did not affect any inflammatory parameters. In contrast, ICAM-1-/- mice treated with anti-VCAM-1 mAb had significantly reduced edema, hemorrhage, and neutrophil infiltration. Furthermore, VCAM-1 blockade in ICAM-1-/- mice suppressed cutaneous TNF-alpha and IL-6 production. Thus, VCAM-1 plays a complementary role to ICAM-1 in the cutaneous Arthus reaction by regulating leukocyte accumulation and proinflammatory cytokine production.


Subject(s)
Arthus Reaction/immunology , Intercellular Adhesion Molecule-1/metabolism , Skin/immunology , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Arthus Reaction/pathology , Edema/immunology , Hemorrhage/immunology , Intercellular Adhesion Molecule-1/biosynthesis , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/immunology , Reverse Transcriptase Polymerase Chain Reaction , Skin/pathology , Tumor Necrosis Factor-alpha/genetics , Vascular Cell Adhesion Molecule-1/biosynthesis
14.
J Invest Dermatol ; 127(2): 447-54, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17008881

ABSTRACT

How neutrophils (polymorphonuclear neutrophils, PMNs) damage vessels in leukocytoclastic vasculitis (LcV) mediated by immune complexes (ICs) is unclear. If degradative enzymes and oxygen radicals are released from PMNs while adhering to the inner side of the vessel wall, they could be washed away by the blood stream or neutralized by serum protease inhibitors. We investigated if in LcV PMNs could damage vessels from the tissue side after transmigration. We used CD18-deficient (CD18-/-) mice because the absence of CD18 excludes transmigration of PMNs. When eliciting the Arthus reaction in ears of CD18-/- mice, deposition of ICs was not sufficient to recruit PMNs or to induce IC-mediated LcV. Injection of PMNs intradermally in CD18-/- mice allowed us to investigate if bypassing diapedesis and placing PMNs exclusively on the abluminal side leads to vascular destruction. We found that injected PMNs gathered around perivascular ICs, but did not cause vessel damage. Only intravenous injection of wild-type PMNs could re-establish the Arthus reaction in CD18-/- mice. Thus, PMNs cause vessel damage during diapedesis from the luminal side, but not from the perivascular space. We suggest that in order to shield the cytotoxic products from the blood stream, ICs induce particularly tight interactions between them, PMNs and endothelial cells.


Subject(s)
Antigen-Antibody Complex/immunology , Blood Vessels/pathology , CD18 Antigens/metabolism , Neutrophil Infiltration , Vasculitis, Leukocytoclastic, Cutaneous/immunology , Vasculitis, Leukocytoclastic, Cutaneous/pathology , Animals , Arthus Reaction/immunology , Arthus Reaction/pathology , CD18 Antigens/genetics , CD18 Antigens/immunology , Cell Adhesion , Cell Degranulation/immunology , Mice , Mice, Knockout/genetics , Neutrophils/immunology , Neutrophils/metabolism , Respiratory Burst
15.
Am J Physiol Heart Circ Physiol ; 291(2): H694-704, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16565304

ABSTRACT

Immune complexes (ICs) are potent inflammatory mediators in peripheral tissues. However, very few studies have examined the ability of ICs to induce inflammatory responses in the brain. Therefore, using preformed ICs or the reverse passive Arthus (RPA) model to localize ICs to the pial microvasculature of mice, we aimed to investigate the ability of ICs to induce an inflammatory response in the cerebral (pial) microvasculature. Application of preformed ICs immediately increased pial microvascular permeability, with a minimal change in leukocyte adhesion in pial postcapillary venules. In contrast, initiation of the RPA response in the pial microvasculature induced changes in cerebral microvascular permeability and increased leukocyte adhesion in pial postcapillary venules. The RPA response induced deposition of C3 in perivascular regions adjacent to sites of IC formation. Depletion of C3 abrogated RPA-induced microvascular permeability and leukocyte adhesion, indicating that the complement pathway was critical for this response. Inhibition of leukocyte adhesion via CD18 blockade also reduced IC-induced microvascular permeability. However, this did not require intercellular adhesion molecule-1, inasmuch as blockade of intercellular adhesion molecule-1 did not alter RPA-induced microvascular permeability and adhesion. These findings demonstrate that ICs are capable of rapidly inducing inflammatory responses in the cerebral microvasculature, with the complement pathway and leukocyte recruitment playing critical roles in microvascular dysfunction.


Subject(s)
Antigen-Antibody Complex/pharmacology , Capillary Permeability/drug effects , Cell Adhesion Molecules/physiology , Complement System Proteins/physiology , Animals , Arthus Reaction/immunology , Arthus Reaction/pathology , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Cerebral Veins/physiology , Complement C3/physiology , Complement System Proteins/metabolism , Endothelial Cells/physiology , Immunohistochemistry , Inflammation/pathology , Leukocyte Count , Male , Mice , Mice, Inbred C57BL , Microcirculation , Ovalbumin/immunology
16.
J Immunol ; 174(5): 3041-50, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15728518

ABSTRACT

Complement and FcgammaR effector pathways are central triggers of immune inflammation; however, the exact mechanisms for their cooperation with effector cells and their nature remain elusive. In this study we show that in the lung Arthus reaction, the initial contact between immune complexes and alveolar macrophages (AM) results in plasma complement-independent C5a production that causes decreased levels of inhibitory FcgammaRIIB, increased levels of activating FcgammaRIII, and highly induced FcgammaR-mediated TNF-alpha and CXCR2 ligand production. Blockade of C5aR completely reversed such changes. Strikingly, studies of pertussis toxin inhibition show the essential role of G(i)-type G protein signaling in C5aR-mediated control of the regulatory FcgammaR system in vitro, and analysis of the various C5aR-, FcgammaR-, and G(i)-deficient mice verifies the importance of Galpha(i2)-associated C5aR and the FcgammaRIII-FcgammaRIIB receptor pair in lung inflammation in vivo. Moreover, adoptive transfer experiments of C5aR- and FcgammaRIII-positive cells into C5aR- and FcgammaRIII-deficient mice establish AM as responsible effector cells. AM lacking either C5aR or FcgammaRIII do not possess any such inducibility of immune complex disease, whereas reconstitution with FcgammaRIIB-negative AM results in an enhanced pathology. These data suggest that AM function as a cellular link of C5a production and C5aR activation that uses a Galpha(i2)-dependent signal for modulating the two opposing FcgammaR, FcgammaRIIB and FcgammaRIII, in the initiation of the inflammatory cascade in the lung Arthus reaction.


Subject(s)
Arthus Reaction/immunology , Arthus Reaction/pathology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Inflammation Mediators/metabolism , Lung/pathology , Macrophages, Alveolar/immunology , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Complement/metabolism , Receptors, Fc/metabolism , Animals , Arthus Reaction/metabolism , Cell Line , Complement C5a/physiology , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Hot Temperature , Immunoglobulin G/pharmacology , Inflammation Mediators/physiology , Lung/immunology , Macrophage Activation/immunology , Macrophages, Alveolar/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/physiology , Receptor Cross-Talk/immunology , Receptor, Anaphylatoxin C5a , Receptors, Complement/biosynthesis , Receptors, Complement/deficiency , Receptors, Complement/genetics , Receptors, IgG/deficiency , Receptors, IgG/genetics , Receptors, IgG/metabolism , Receptors, IgG/physiology
17.
Folia Histochem Cytobiol ; 42(2): 101-10, 2004.
Article in English | MEDLINE | ID: mdl-15253133

ABSTRACT

Supramolecular micellar structures have been proposed as carriers in aim-oriented drug transportation to a target marked by specific immune complexes. In this study, the self-assembling dye Congo red was used as a model supramolecular carrier and its accumulation in the target was studied in vivo. The target was created in vivo as the local specific inflammation provoked by subcutaneous injection of antigen to the ear of a previously immunized rabbit. The color caused by accumulation of Congo red after its intravenous injection was registered by pictures of the ear with suitably filtered visible light shining through it to distinguish Congo red against the background color of hemoglobin. The results confirmed the expected accumulation and retention of Congo red in the inflammation area marked by deposits of specific immune complexes. The role of albumin and its possible interference with transportation of drugs through the blood by supramolecular carriers was also subjected to preliminary examination. The results revealed that albumin collaborates rather than interferes with drug transportation; this is another factor making the use of supramolecular carriers for aim-oriented chemotherapy highly promising.


Subject(s)
Antigen-Antibody Complex/metabolism , Congo Red/metabolism , Congo Red/pharmacokinetics , Ear/physiopathology , Albumins/chemistry , Albumins/metabolism , Animals , Arthus Reaction/metabolism , Arthus Reaction/pathology , Arthus Reaction/physiopathology , Coloring Agents/chemistry , Coloring Agents/metabolism , Coloring Agents/pharmacokinetics , Congo Red/chemistry , Drug Therapy/methods , Immunohistochemistry , Kinetics , Models, Molecular , Molecular Structure , Rabbits , Rhodamines/chemistry , Rhodamines/metabolism
18.
J Immunol ; 170(8): 4310-7, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12682267

ABSTRACT

Complement C2 receptor inhibitor trispanning (CRIT) is a Schistosoma protein that binds the human complement protein, C2. We recently showed that peptides based on the ligand binding region of CRIT inhibit the classical pathway (CP) of complement activation in human serum, using hemolytic assays and so speculated that on the parasite surface CRIT has the function of evading human complement. We now show that in vitro the C2-binding 11-aa C terminus of the first extracellular domain of CRIT, a 1.3-kDa peptide termed CRIT-H17, inhibits CP activation in a species-specific manner, inhibiting mouse and rat complement but not that from guinea pig. Hitherto, the ability of CRIT to regulate complement in vivo has not been assessed. In this study we show that by inhibiting the CP, CRIT-H17 is able to reduce immune complex-mediated inflammation (dermal reversed passive Arthus reaction) in BALB/c mice. Upon intradermal injection of CRIT-H17, and similarly with recombinant soluble complement receptor type 1, there was a 41% reduction in edema and hemorrhage, a 72% reduction in neutrophil influx, and a reduced C3 deposition. Furthermore, when H17 was administered i.v. at a 1 mg/kg dose, inflammation was reduced by 31%. We propose that CRIT-H17 is a potential therapeutic agent against CP complement-mediated inflammatory tissue destruction.


Subject(s)
Antigen-Antibody Complex/pharmacology , Antigens, Helminth , Antigens, Protozoan/pharmacology , Complement Inactivator Proteins/pharmacology , Helminth Proteins , Immunosuppressive Agents/pharmacology , Peptide Fragments/pharmacology , Protozoan Proteins/pharmacology , Receptors, Complement 3d/antagonists & inhibitors , Skin/immunology , Skin/pathology , Animals , Antigens, Protozoan/administration & dosage , Arthus Reaction/immunology , Arthus Reaction/metabolism , Arthus Reaction/pathology , Complement C3/metabolism , Complement Inactivator Proteins/administration & dosage , Ear, External , Female , Guinea Pigs , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/prevention & control , Injections, Intravenous , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/immunology , Peptide Fragments/administration & dosage , Protozoan Proteins/administration & dosage , Rats , Receptors, Cell Surface/administration & dosage , Receptors, Complement 3d/metabolism , Schistosoma/immunology , Species Specificity , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
19.
Am J Pathol ; 162(5): 1463-73, 2003 May.
Article in English | MEDLINE | ID: mdl-12707029

ABSTRACT

Immune complex-induced tissue injury is mediated by inflammatory cell infiltration that is highly regulated by multiple adhesion molecules. To assess the relative contribution of adhesion molecules, including selectins and ICAM-1, in this pathogenetic process, the cutaneous passive Arthus reaction was examined in mice lacking E-selectin, P-selectin, or both L-selectin and ICAM-1 with anti-P- or E-selectin mAbs. Edema and hemorrhage were significantly reduced in P-selectin(-/-) mice compared with wild-type mice while they were not inhibited in E-selectin(-/-) mice. Combined E- and P-selectin blockade resulted in more significant reduction relative to L-selectin/ICAM-1(-/-) as well as P-selectin(-/-) mice. Remarkably, both E- and P-selectin blockade in L-selectin/ICAM-1(-/-) mice completely abrogated edema and hemorrhage. The inhibited edema and hemorrhage paralleled reduced infiltration of neutrophils and mast cells that expressed significant levels of P-selectin glycoprotein ligand-1. Similarly reduced infiltration of neutrophils and mast cells was observed in the peritoneal Arthus reaction and was associated partly with the decreased production of tumor necrosis factor-alpha and interleukin-6. The results of this study indicate that both endothelial selectins contribute predominantly to the Arthus reaction by regulating mast cell and neutrophil infiltration and that the full development of the Arthus reaction is mediated cooperatively by all selectins and ICAM-1.


Subject(s)
E-Selectin/physiology , Immune Complex Diseases/pathology , Intercellular Adhesion Molecule-1/physiology , P-Selectin/physiology , Selectins/physiology , Animals , Arthus Reaction/genetics , Arthus Reaction/pathology , Arthus Reaction/physiopathology , Cell Adhesion Molecules/physiology , E-Selectin/genetics , Edema/genetics , Edema/pathology , Edema/physiopathology , Hemorrhage/genetics , Hemorrhage/pathology , Hemorrhage/physiopathology , Inflammation/pathology , Inflammation/physiopathology , Intercellular Adhesion Molecule-1/genetics , Mice , Mice, Knockout , P-Selectin/genetics
20.
J Allergy Clin Immunol ; 110(3): 413-20, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12209088

ABSTRACT

BACKGROUND: Immunotherapy has gradually fallen out of favor for the treatment of many allergic diseases because of the overall convenience, safety, and efficacy of medications. However, investigations suggest that allergen/immunostimulatory sequence oligodeoxynucleotide (ISS-ODN) conjugates (AICs) might have improved safety and efficacy compared with allergen extracts. OBJECTIVE: We determined whether changes in the ISS-ODN conjugation ratio would effect the immunogenicity and allergenicity of AIC. METHODS: Immunogenicity was determined by means of AIC vaccination of mice, followed by analysis of antigen-specific antibody and cytokine responses. The allergenicity of AIC was determined in mast cell release studies and in murine models of anaphylaxis and the Arthus reaction. RESULTS: AIC induced a stronger immune response than allergen alone or allergen mixed with ISS-ODN, but higher-level ISS-ODN conjugation reduced its immunogenicity modestly. In mast cell degranulation studies AIC was approximately 100-fold less allergenic than native allergen, with stepwise increases in the ODN conjugation ratio leading to stepwise decreases in allergenicity. In anaphylaxis studies death rates were reduced from 100% with native allergen challenge to as low as 0% with high-ratio ISS-ODN AIC challenge. Similar results were obtained in an Arthus reaction model. CONCLUSION: These investigations establish that AIC is both significantly more immunogenic and less allergenic than native allergens and the techniques used might have further utility for the standardization and optimization of AIC formulations for use in allergic patients.


Subject(s)
Adjuvants, Immunologic , Allergens/immunology , Anaphylaxis/etiology , Oligodeoxyribonucleotides , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Allergens/chemistry , Anaphylaxis/mortality , Anaphylaxis/prevention & control , Animal Population Groups , Animals , Arthus Reaction/etiology , Arthus Reaction/pathology , Binding Sites, Antibody , Cell Degranulation , Cells, Cultured , Epitopes/immunology , Female , Immunotherapy/methods , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/adverse effects , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/pharmacology , Ovalbumin/immunology , Rats , Survival Analysis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...