Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38862755

ABSTRACT

Allergic disease is a major global health concern that imposes significant life-altering and economic burdens on affected individuals. However, there is still no cure. Polymer-based nanoparticles (NP) have shown the potential to induce antigen (Ag)-specific immune tolerance in various Th1/17 and Th2-mediated immune disorders including autoimmunity and allergy. Common methods by which Ags are associated with NPs are through surface conjugation or encapsulation. However, these Ag delivery strategies can be associated with several caveats that dampen their effectiveness such as uncontrolled Ag loading, a high Ag burst release, and an increased immune recognition profile. We previously developed Ag-polymer conjugate NPs (acNPs) to overcome those noted limitations, while allowing for controlled delivery of precise quantities of Ag to innate immune cells for Ag-specific CD4 T cell modulation. Here, we utilized ovalbumin (OVA) protein-poly(lactic-co-glycolic acid) (PLGA) conjugate NPs (acNP-OVA) to elucidate the impact of Ag loading on the induction of Th2 tolerance using a prophylactic and therapeutic OVA/ALUM-induced mouse model of allergic lung inflammation (ALI) in comparison to Ag-encapsulated PLGA NPs (NP(Ag)). We demonstrate that acNP-OVA formulations reduced OVA-specific IgE and inhibited Th2 cytokine secretions in an Ag loading-dependent manner when administered prophylactically. Administration of acNP-OVA to pre-sensitized mice did not affect OVA-specific IgE and Th2 cytokines tended to be reduced, however, there was no clear Ag loading dependency. acNP-OVA with medium-to-low Ag loadings were well tolerated, while formulations with high Ag loadings, including NP(Ag) resulted in anaphylaxis. Overall, our results clarify the relationship between Ag loading and Ag-specific IgE and Th2 cytokine responses in a murine model of ALI, which provides insight useful for future design of tolerogenic NP-based immunotherapies.

2.
Front Immunol ; 14: 1297354, 2023.
Article in English | MEDLINE | ID: mdl-38259471

ABSTRACT

We investigated the effect of global Plexin B1 deficiency on allergic airway responses to house dust mite (HDM) or ovalbumin (OVA). In the HDM model, there were higher Th2 cytokine levels in the BALF of Plexin B1 knock-out (KO) mice compared to wild type (WT), and tissue inflammation and mucus production were modestly enhanced. In the OVA model, Plexin B1 deficiency led to increases in lung inflammation, mucus production, and lung Th2 cytokines accompanied by dysregulated mucin gene expression without affecting anti-OVA IgE/IgG1 levels. Spleen cells from Plexin B1 KO mice proliferated more robustly than WT cells in vitro to a variety of stimuli. Plexin B1 KO CD4+ T cells from spleens expressed higher levels of Ki-67 and CD69 compared to WT cells. Spleen cells from naïve Plexin B1 KO mice secreted increased amounts of IL-4 and IL-6 when pulsed in vitro with OVA whereas in vivo OVA-primed spleen cells produced IL-4/IL-5 when subjected to in vitro OVA restimulation. The upregulated allergic inflammatory response in Plexin B1 KO mice was associated with a lower number of Tregs in the lung tissues. Moreover, these mice displayed lower numbers of Treg cells in the lymphoid tissues at the baseline. These results demonstrate a previously unrecognized link between Plexin B1, Treg cells, and mucus in allergic lung inflammation.


Subject(s)
Hypersensitivity , Nerve Tissue Proteins , Receptors, Cell Surface , T-Lymphocytes, Regulatory , Animals , Mice , Cytokines , Dermatophagoides pteronyssinus , Inflammation , Interleukin-4 , Mucins , Nerve Tissue Proteins/metabolism , Ovalbumin , Pneumonia , Receptors, Cell Surface/metabolism
3.
J Immunol ; 208(9): 2085-2097, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35396219

ABSTRACT

Asthma is a common and ubiquitous chronic respiratory disease that is associated with airway inflammation and hyperreactivity resulting in airway obstruction. It is now accepted that asthma is controlled by a combination of host genetics and environment in a rather complex fashion; however, the link between sensing of the environment and development and exacerbation of allergic lung inflammation is unclear. Human populations expressing cosegregating D299G and T399I polymorphisms in the TLR4 gene are associated with a decreased risk for asthma in adults along with hyporesponsiveness to inhaled LPS, the TLR4 ligand. However, these data do not account for other human genetic or environmental factors. Using a novel mouse strain that expresses homologous human TLR4 polymorphisms (TLR4-single nucleotide polymorphism [SNP]), we directly tested the effect of these TLR4 polymorphisms on in vivo responses to allergens using two models of induction. We report that intact TLR4 is required for allergic inflammation when using the OVA and LPS model of induction, as cellular and pathological benchmarks were diminished in both TLR4-SNP and TLR4-deficent mice. However, in the more clinically relevant model using house dust mite extract for induction, responses were enhanced in the TLR4-SNP mice, as evidenced by greater levels of eosinophilic inflammation, Th2 cytokine production, and house dust mite-specific IgG1 production compared with wild-type mice; however, mucus production and airway hyperreactivity were not affected. These results suggest that the TLR4 polymorphic variants (genes) interact differently with the allergic stimulation (environment).


Subject(s)
Antigens, Dermatophagoides , Asthma , Pulmonary Eosinophilia , Toll-Like Receptor 4 , Allergens , Animals , Antigens, Dermatophagoides/immunology , Asthma/genetics , Asthma/pathology , Inflammation , Lipopolysaccharides , Mice , Polymorphism, Single Nucleotide , Pyroglyphidae , Toll-Like Receptor 4/genetics
4.
Int J Mol Sci ; 23(6)2022 Mar 11.
Article in English | MEDLINE | ID: mdl-35328445

ABSTRACT

Semaphorin 4A (Sema4A) exerts a stabilizing effect on human Treg cells in PBMC and CD4+ T cell cultures by engaging Plexin B1. Sema4A deficient mice display enhanced allergic airway inflammation accompanied by fewer Treg cells, while Sema4D deficient mice displayed reduced inflammation and increased Treg cell numbers even though both Sema4 subfamily members engage Plexin B1. The main objectives of this study were: 1. To compare the in vitro effects of Sema4A and Sema4D proteins on human Treg cells; and 2. To identify function-determining residues in Sema4A critical for binding to Plexin B1 based on Sema4D homology modeling. We report here that Sema4A and Sema4D display opposite effects on human Treg cells in in vitro PBMC cultures; Sema4D inhibited the CD4+CD25+Foxp3+ cell numbers and CD25/Foxp3 expression. Sema4A and Sema4D competitively bind to Plexin B1 in vitro and hence may be doing so in vivo as well. Bayesian Partitioning with Pattern Selection (BPPS) partitioned 4505 Sema domains from diverse organisms into subgroups based on distinguishing sequence patterns that are likely responsible for functional differences. BPPS groups Sema3 and Sema4 into one family and further separates Sema4A and Sema4D into distinct subfamilies. Residues distinctive of the Sema3,4 family and of Sema4A (and by homology of Sema4D) tend to cluster around the Plexin B1 binding site. This suggests that the residues both common to and distinctive of Sema4A and Sema4D may mediate binding to Plexin B1, with subfamily residues mediating functional specificity. We mutated the Sema4A-specific residues M198 and F223 to alanine; notably, F223 in Sema4A corresponds to alanine in Sema4D. Mutant proteins were assayed for Plexin B1-binding and Treg stimulation activities. The F223A mutant was unable to stimulate Treg stability in in vitro PBMC cultures despite binding Plexin B1 with an affinity similar to the WT protein. This research is a first step in generating potent mutant Sema4A molecules with stimulatory function for Treg cells with a view to designing immunotherapeutics for asthma.


Subject(s)
Leukocytes, Mononuclear , Semaphorins/metabolism , Alanine , Animals , Bayes Theorem , Forkhead Transcription Factors/genetics , Humans , Inflammation , Leukocytes, Mononuclear/metabolism , Mice , Nerve Tissue Proteins/metabolism
5.
Mol Med ; 27(1): 162, 2021 12 27.
Article in English | MEDLINE | ID: mdl-34961486

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel type b coronavirus responsible for the COVID-19 pandemic. With over 224 million confirmed infections with this virus and more than 4.6 million people dead because of it, it is critically important to define the immunological processes occurring in the human response to this virus and pathogenetic mechanisms of its deadly manifestation. This perspective focuses on the contribution of the recently discovered interaction of SARS-CoV-2 Spike protein with neuropilin 1 (NRP1) receptor, NRP1 as a virus entry receptor for SARS-CoV-2, its role in different physiologic and pathologic conditions, and the potential to target the Spike-NRP1 interaction to combat virus infectivity and severe disease manifestations.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Neuropilin-1/chemistry , Neuropilin-1/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/epidemiology , COVID-19/etiology , Comorbidity , Diabetes Mellitus/epidemiology , Diabetes Mellitus/virology , Female , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Infant , Molecular Targeted Therapy/methods , Neuropilin-1/immunology , Pregnancy , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/virology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism
6.
Immunohorizons ; 3(2): 71-87, 2019 02.
Article in English | MEDLINE | ID: mdl-31236543

ABSTRACT

We previously reported that neuroimmune semaphorin (Sema) 4A regulates the severity of experimental allergic asthma and increases regulatory T (Treg) cell numbers in vivo; however, the mechanisms of Sema4A action remain unknown. It was also reported that Sema4A controls murine Treg cell function and survival acting through neuropilin 1 (NRP-1) receptor. To clarify Sema4A action on human T cells, we employed T cell lines (HuT78 and HuT102), human PBMCs, and CD4+ T cells in phenotypic and functional assays. We found that HuT78 demonstrated a T effector-like phenotype (CD4+CD25lowFoxp3-), whereas HuT102 expressed a Treg-like phenotype (CD4+CD25hi Foxp3+). Neither cell line expressed NRP-1. HuT102 cells expressed Sema4A counter receptor Plexin B1, whereas HuT78 cells were Sema4A+. All human peripheral blood CD4+ T cells, including Treg cells, expressed PlexinB1 and lacked both NRP-1 and -2. However, NRP-1 and Sema4A were detected on CD3negativeCD4intermediate human monocytes. Culture of HuT cells with soluble Sema4A led to an upregulation of CD25 and Foxp3 markers on HuT102 cells. Addition of Sema4A increased the relative numbers of CD4+CD25+Foxp3+ cells in PBMCs and CD4+ T cells, which were NRP-1negative but PlexinB1+, suggesting the role of this receptor in Treg cell stability. The inclusion of anti-PlexinB1 blocking Ab in cultures before recombinant Sema4A addition significantly decreased Treg cell numbers as compared with cultures with recombinant Sema4A alone. Sema4A was as effective as TGF-ß in inducible Treg cell induction from CD4+CD25depleted cells but did not enhance Treg cell suppressive activity in vitro. These results suggest strategies for the development of new Sema4A-based therapeutic measures to combat allergic inflammatory diseases. ImmunoHorizons, 2019, 3: 71-87.


Subject(s)
Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Semaphorins/pharmacology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Antibodies , Asthma/immunology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Forkhead Transcription Factors/metabolism , Humans , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Nerve Tissue Proteins/immunology , Neuropilin-1/metabolism , Phenotype , Receptors, Cell Surface/immunology , Transforming Growth Factor beta/metabolism
7.
Mol Med ; 24(1): 13, 2018 04 04.
Article in English | MEDLINE | ID: mdl-30134791

ABSTRACT

Several neuronal guidance proteins, known as semaphorin molecules, function in the immune system. This dual tissue performance has led to them being defined as "neuroimmune semaphorins". They have been shown to regulate T cell activation by serving as costimulatory molecules. Similar to classical costimulatory molecules, neuroimmune semaphorins are either constitutively or inducibly expressed on immune cells. In contrast to the classical costimulatory molecule function, the action of neuroimmune semaphorins requires the presence of two signals, the first one provided by TCR/MHC engagement, and the second one provided by B7/CD28 interaction. Thus, neuroimmune semaphorins serve as a "signal three" for immune cell activation and regulate the overall intensity of immune response. The current knowledge on their structures, multiple receptors, specific cell/tissue/organ expression, and distinct functions in different diseases are summarized and discussed in this review.


Subject(s)
Semaphorins/immunology , Animals , Humans , Nervous System/immunology , Semaphorins/chemistry , Semaphorins/metabolism
8.
Int J Mol Sci ; 20(1)2018 Dec 30.
Article in English | MEDLINE | ID: mdl-30598022

ABSTRACT

Neuroimmune semaphorin 4A (Sema4A), a member of semaphorin family of transmembrane and secreted proteins, is an important regulator of neuronal and immune functions. In the nervous system, Sema4A primarily regulates the functional activity of neurons serving as an axon guidance molecule. In the immune system, Sema4A regulates immune cell activation and function, instructing a fine tuning of the immune response. Recent studies have shown a dysregulation of Sema4A expression in several types of cancer such as hepatocellular carcinoma, colorectal, and breast cancers. Cancers have been associated with abnormal angiogenesis. The function of Sema4A in angiogenesis and cancer is not defined. Recent studies have demonstrated Sema4A expression and function in endothelial cells. However, the results of these studies are controversial as they report either pro- or anti-angiogenic Sema4A effects depending on the experimental settings. In this mini-review, we discuss these findings as well as our data on Sema4A regulation of inflammation and angiogenesis, which both are important pathologic processes underlining tumorigenesis and tumor metastasis. Understanding the role of Sema4A in those processes may guide the development of improved therapeutic treatments for cancer.


Subject(s)
Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Semaphorins/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinogenesis/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Semaphorins/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
Inflamm Res ; 66(2): 111-117, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27554682

ABSTRACT

Long-term inflammatory processes directed at a particular endogenous or exogenous antigen, or sometimes of unknown etiology, form the pathogenetic basis for many debilitating conditions, such as cardiovascular, pulmonary, autoimmune, neurologic diseases, and cancer. Recent discoveries of neuroimmune semaphorins 4A and 4D (Sema4A and Sema4D, respectively) expression and function in the immune system and their key regulatory roles in fine tuning of inflammatory processes made them the molecules of interest for a potential immunotherapy. In this short review, we discuss the current knowledge in the Sema4A and Sema4D actions in chronic inflammation underlying the outlined above diseases.


Subject(s)
Antigens, CD/immunology , Semaphorins/immunology , Animals , Arthritis, Rheumatoid/immunology , Asthma/immunology , Atherosclerosis/immunology , Humans , Inflammatory Bowel Diseases/immunology , Lupus Erythematosus, Systemic/immunology , Multiple Sclerosis/immunology , Myocarditis/immunology , Neoplasms/immunology , Nerve Tissue Proteins/immunology , Receptors, Cell Surface/immunology
10.
Arthritis Res Ther ; 17: 313, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26542940

ABSTRACT

Rheumatoid arthritis (RA) is a systemic autoimmune disease manifesting in joint destruction. The recognized hallmark of RA pathogenesis is the involvement of immune cells which produce many mediators potentiating an inflammatory environment. RA synovial fibroblasts (RASFs) contribute significantly to disease progression by initiating and regulating many pathways of joint destruction. Detailed molecular insights into RASF biology may lead to identification of important therapeutic targets. The discovery of common molecular targets for joint resident and inflammatory cells may help to develop the most effective therapeutic strategy. One such pathway includes semaphorin 4A as reported in a recent article in Arthritis Research & Therapy.


Subject(s)
Arthritis, Rheumatoid/genetics , Fibroblasts/metabolism , Gene Expression/genetics , Semaphorins/genetics , Synovial Membrane/metabolism , Female , Humans , Male
11.
J Immunol ; 191(4): 1517-28, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23825312

ABSTRACT

STAT6 plays a central role in IL-4-mediated allergic responses. Several studies indicate that regulatory T cells (Tregs) can be modulated by IL-4 in vitro. We previously showed that STAT6(-/-) mice are highly resistant to allergic lung inflammation even when wild-type Th2 effectors were provided and that they have increased numbers of Tregs. However, the role of STAT6 in modulating Tregs in vivo during allergic lung inflammation has not been thoroughly investigated. To examine Treg and STAT6 interaction during allergic inflammation, STAT6(-/-), STAT6xRAG2(-/-), and RAG2(-/-) mice were subjected to OVA sensitization and challenge following adoptive transfer of OVA-specific, wild-type Th2 effectors with or without prior Treg depletion/inactivation, using anti-CD25 (PC61). As expected, STAT6(-/-) mice were highly resistant to airway inflammation and remodeling. In contrast, allergic lung inflammation was partially restored in STAT6(-/-) mice treated with PC61 to levels observed in STAT6xRAG2(-/-) mice. In some cases, STAT6xRAG2(-/-) mice were also given natural Tregs along with Th2 effectors. Adoptive transfer of natural Tregs caused a substantial reduction in bronchoalveolar lavage eosinophil composition and suppressed airway remodeling and T cell migration into the lung in STAT6xRAG2(-/-) mice to levels comparable to those in STAT6(-/-) mice. These results demonstrate the STAT6-dependent suppression of Tregs in vivo to promote allergic airway inflammation.


Subject(s)
Pulmonary Eosinophilia/immunology , STAT6 Transcription Factor/physiology , T-Lymphocytes, Regulatory/immunology , Administration, Intranasal , Adoptive Transfer , Airway Remodeling , Allergens/administration & dosage , Allergens/toxicity , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , DNA-Binding Proteins/deficiency , Forkhead Transcription Factors/analysis , Immune Tolerance , Interleukin-2 Receptor alpha Subunit/analysis , Interleukin-5/analysis , Lung/immunology , Lung/pathology , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/toxicity , Pulmonary Eosinophilia/etiology , Pulmonary Eosinophilia/pathology , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/genetics , T-Lymphocytes, Regulatory/classification , T-Lymphocytes, Regulatory/transplantation , Th2 Cells/immunology
12.
F1000 Biol Rep ; 5: 3, 2013.
Article in English | MEDLINE | ID: mdl-23413371

ABSTRACT

Cytokines are small, secreted proteins that control immune responses. Within the lung, they can control host responses to injuries or infection, resulting in clearance of the insult, repair of lung tissue, and return to homeostasis. Problems can arise when this response is over exuberant and/or cytokine production becomes dysregulated. In such cases, chronic and repeated inflammatory reactions and cytokine production can be established, leading to airway remodeling and fibrosis with unintended, maladaptive consequences. In this report, we describe the cytokines and molecular mechanisms behind the pathology observed in three major chronic diseases of the lung: asthma, chronic obstructive pulmonary disease (COPD), and pulmonary fibrosis. Overlapping mechanisms are presented as potential sites for therapeutic intervention.

13.
BMC Immunol ; 12: 60, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-22014099

ABSTRACT

BACKGROUND: CD4+ T helper type 2 (TH2) cells, their cytokines IL-4, IL-5 and IL-13 and the transcription factor STAT6 are known to regulate various features of asthma including lung inflammation, mucus production and airway hyperreactivity and also drive alternative activation of macrophages (AAM). However, the precise roles played by the IL-4/IL-13 receptors and STAT6 in inducing AAM protein expression and modulating specific features of airway inflammation are still unclear. Since TH2 differentiation and activation plays a pivotal role in this disease, we explored the possibility of developing an asthma model in mice using T cells that were differentiated in vivo. RESULTS: In this study, we monitored the activation and proliferation status of adoptively transferred allergen-specific naïve or in vivo primed CD4+ T cells. We found that both the naïve and in vivo primed T cells expressed similar levels of CD44 and IL-4. However, in vivo primed T cells underwent reduced proliferation in a lymphopenic environment when compared to naïve T cells. We then used these in vivo generated effector T cells in an asthma model. Although there was reduced inflammation in mice lacking IL-4Rα or STAT6, significant amounts of eosinophils were still present in the BAL and lung tissue. Moreover, specific AAM proteins YM1 and FIZZ1 were expressed by epithelial cells, while macrophages expressed only YM1 in RAG2-/- mice. We further show that FIZZ1 and YM1 protein expression in the lung was completely dependent on signaling through the IL-4Rα and STAT6. Consistent with the enhanced inflammation and AAM protein expression, there was a significant increase in collagen deposition and smooth muscle thickening in RAG2-/- mice compared to mice deficient in IL-4Rα or STAT6. CONCLUSIONS: These results establish that transfer of in vivo primed CD4+ T cells can induce allergic lung inflammation. Furthermore, while IL-4/IL-13 signaling through IL-4Rα and STAT6 is essential for AAM protein expression, lung inflammation and eosinophilia are only partially dependent on this pathway. Further studies are required to identify other proteins and signaling pathways involved in airway inflammation.


Subject(s)
Asthma/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Lectins/metabolism , Pneumonia/immunology , Th2 Cells/metabolism , beta-N-Acetylhexosaminidases/metabolism , Adoptive Transfer , Animals , Cells, Cultured , Complement Pathway, Alternative/genetics , Disease Models, Animal , Hyaluronan Receptors/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Lectins/genetics , Lectins/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Pneumonia/genetics , Pulmonary Eosinophilia/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction/immunology , Th2 Cells/immunology , Th2 Cells/pathology , Th2 Cells/transplantation , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/immunology
14.
BMC Immunol ; 12: 30, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21595947

ABSTRACT

BACKGROUND: Semaphorins were originally identified as molecules regulating a functional activity of axons in the nervous system. Sema4A and Sema4D were the first semaphorins found to be expressed on immune cells and were termed "immune semaphorins". It is known that Sema4A and Sema4D bind Tim-2 and CD72 expressed on leukocytes and PlexinD1 and B1 present on non-immune cells. These neuroimmune semaphorins and their receptors have been shown to play critical roles in many physiological and pathological processes including neuronal development, immune response regulation, cancer, autoimmune, cardiovascular, renal, and infectious diseases. However, the expression and regulation of Sema4A, Sema4D, and their receptors in normal and allergic lungs is undefined. RESULTS: Allergen treatment and lung-specific vascular endothelial growth factor (VEGF) expression induced asthma-like pathologies in the murine lungs. These experimental models of allergic airway inflammation were used for the expression analysis of immune semaphorins and their receptors employing immunohistochemistry and flow cytometry techniques. We found that besides accessory-like cells, Sema4A was also detected on bronchial epithelial and smooth muscle cells, whereas Sema4D expression was high on immune cells such as T and B lymphocytes. Surprisingly, under inflammation various cell types including macrophages, lymphocytes, and granulocytes in the lung expressed Tim-2, a previously defined marker for Th2 cells. CD72 was found on lung immune, inflammatory, and epithelial cells. Bronchial epithelial cells were positive for both plexins, whereas some endothelial cells selectively expressed Plexin D1. Plexin B1 expression was also detected on lung DC. Both allergen and VEGF upregulated the expression of neuroimmune semaphorins and their receptors in the lung tissue. However, the lung tissue Sema4A-Tim2 expression was rather weak, whereas Sema4D-CD72 ligand-receptor pair was vastly upregulated by allergen. Soluble Sema4D protein was present in the lung lysates and a whole Sema4A protein plus its dimer were readily detected in the bronchoalveolar (BAL) fluids under inflammation. CONCLUSIONS: This study clearly shows that neuroimmune semaphorins Sema4A and Sema4D and their receptors might serve as potential markers for the allergic airway inflammatory diseases. Our current findings pave the way for further investigations of the role of immune semaphorins in inflammation and their use as potential therapeutic targets for the inflammatory lung conditions.


Subject(s)
Asthma/immunology , Lung/metabolism , Pneumonia/immunology , Semaphorins/metabolism , Th2 Cells/metabolism , Allergens/administration & dosage , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Humans , Immunohistochemistry , Lung/drug effects , Lung/immunology , Lung/pathology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Ovalbumin/administration & dosage , Pneumonia/chemically induced , Semaphorins/genetics , Semaphorins/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/pathology , Up-Regulation/drug effects , Up-Regulation/immunology , Vascular Endothelial Growth Factor A/administration & dosage
15.
Immunol Res ; 50(1): 10-21, 2011 May.
Article in English | MEDLINE | ID: mdl-21203905

ABSTRACT

Semaphorins belong to a family of membrane-bound and secreted molecules that regulate the functional activity of axons in the nervous system. Sema4A and Sema4D were the first semaphorins also found to be expressed in immune cells and were, therefore, termed "immune semaphorins". It is known that Sema4A has three functional receptors, namely Plexin D1, Plexin B1, and Tim-2, whereas Sema4D binds to Plexin B1 and CD72. Recent studies suggest that immune semaphorins play critical roles in many physiological and pathological processes and such. In this review, we summarize the current knowledge on the biology of neuroimmune semaphorins and their corresponding receptors, their distribution in organs and tissues, function in the immune response, and critical regulatory roles in various diseases.


Subject(s)
Neuroimmunomodulation/immunology , Semaphorins/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/immunology , Disease , Humans , Immunity/immunology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/immunology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/immunology , Semaphorins/chemistry
16.
J Immunol ; 186(4): 2571-83, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21242523

ABSTRACT

Th2 cells induce asthma through the secretion of cytokines. Two such cytokines, IL-4 and IL-13, are critical mediators of many features of this disease. They both share a common receptor subunit, IL-4Rα, and signal through the STAT6 pathway. STAT6(-/-) mice have impaired Th2 differentiation and reduced airway response to allergen. Transferred Th2 cells were not able to elicit eosinophilia in response to OVA in STAT6(-/-) mice. To clarify the role of STAT6 in allergic airway inflammation, we generated mouse bone marrow (BM) chimeras. We observed little to no eosinophilia in OVA-treated STAT6(-/-) mice even when STAT6(+/+) BM or Th2 cells were provided. However, when Th2 cells were transferred to STAT6×Rag2(-/-) mice, we observed an eosinophilic response to OVA. Nevertheless, the expression of STAT6 on either BM-derived cells or lung resident cells enhanced the severity of OVA-induced eosinophilia. Moreover, when both the BM donor and recipient lacked lymphocytes, transferred Th2 cells were sufficient to induce the level of eosinophilia comparable with that of wild-type (WT) mice. The expression of STAT6 in BM-derived cells was more critical for the enhanced eosinophilic response. Furthermore, we found a significantly higher number of CD4(+)CD25(+)Foxp3(+) T cells (regulatory T cells [Tregs]) in PBS- and OVA-treated STAT6(-/-) mouse lungs compared with that in WT animals suggesting that STAT6 limits both naturally occurring and Ag-induced Tregs. Tregs obtained from either WT or STAT6(-/-) mice were equally efficient in suppressing CD4(+) T cell proliferation in vitro. Taken together, our studies demonstrate multiple STAT6-dependent and -independent features of allergic inflammation, which may impact treatments targeting STAT6.


Subject(s)
Gene Expression Regulation/immunology , Ovalbumin/immunology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , STAT6 Transcription Factor/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cells, Cultured , Coculture Techniques , Immunophenotyping , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lung/immunology , Lung/pathology , Lymphocyte Cooperation/genetics , Lymphocyte Cooperation/immunology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Respiratory Hypersensitivity/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/genetics , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Th2 Cells/transplantation
17.
J Neuroimmunol ; 225(1-2): 82-90, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20605223

ABSTRACT

The chemokine CCL27 has chemoattractant properties for memory T cells and has been implicated in skin allergic reactions. The present study reports the expression in the brain of two CCL27 splice variants localized in the cerebral cortex and limbic regions. CCL27-like immunoreactivity was identified mainly in neurons. Variant 1 was found elevated in the olfactory bulbs during allergic inflammation induced by intranasal challenge with allergen. This was accompanied by the presence of T cells in the olfactory bulbs. Intranasal administration of neutralizing antibodies against CCL27 reduced the presence of T cells in the olfactory bulbs suggesting a function in T cell activity in the brain.


Subject(s)
Brain/metabolism , Chemokine CCL27/genetics , Chemokine CCL27/metabolism , Gene Expression Regulation/immunology , Albumins/adverse effects , Albumins/immunology , Analysis of Variance , Animals , Autoradiography/methods , Brain/anatomy & histology , Brain/cytology , CD3 Complex/metabolism , Cell Count/methods , Hypersensitivity/immunology , Hypersensitivity/metabolism , Male , Mice , Mice, Inbred BALB C , Neurons/metabolism , Protein Isoforms/genetics , RNA, Messenger/metabolism
18.
J Leukoc Biol ; 87(6): 1011-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20335310

ABSTRACT

During the development of immune responses to pathogens, self-antigens, or environmental allergens, naive CD4(+) T cells differentiate into subsets of effector cells including Th1, Th2, and Th17 cells. The differentiation into these subsets is controlled by specific transcription factors. The activity of these effector cells is limited by nTregs and iTregs, whose differentiation and maintenance are dependent on the transcription factor Foxp3. The regulation of autoimmune diseases mediated by Th1 and Th17 cells by Tregs has been studied and reviewed extensively. However, much less has been presented about the interplay between Tregs and Th2 cells and their contribution to allergic disease. In this perspective, we discuss the regulation of Th2 cells by Tregs and vice versa, focusing on the interplay between the IL-4-activated STAT6/GATA3 pathway and Foxp3.


Subject(s)
Interleukin-4/metabolism , STAT6 Transcription Factor/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Animals , Humans
19.
Clin Immunol ; 132(3): 371-84, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19553159

ABSTRACT

We previously demonstrated that vascular endothelial growth factor (VEGF) expression in the murine lung increases local CD11c+MHCII+ DC number and activation. In this study, employing a multicolor flow cytometry, we report increases in both myeloid (mDC) and plasmacytoid (pDC) DC in the lungs of VEGF transgenic (tg) compared to WT mice. Lung pDC from VEGF tg mice exhibited higher levels of activation with increased expression of MHCII and costimulatory molecules. As VEGF tg mice display an asthma-like phenotype and lung mDC play a critical role in asthmatic setting, studies were undertaken to further characterize murine lung mDC. Evaluations of sorted mDC from VEGF tg lungs demonstrated a selective upregulation of cathepsin K, MMP-8, -9, -12, and -14, and chemokine receptors as compared to those obtained from WT control mice. They also had increased VEGFR2 but downregulated VEGFR1 expression. Analysis of chemokine and regulatory cytokine expression in these cells showed an upregulation of macrophage chemotactic protein-3 (MCP-3), thymus-expressed chemokine (TECK), secondary lymphoid organ chemokine (SLC), macrophage-derived chemokine (MDC), IL-1beta, IL-6, IL-12 and IL-13. The antigen (Ag) OVA-FITC uptake by lung DC and the migration of Ag-loaded DC to local lymph nodes were significantly increased in VEGF tg mice compared to WT mice. Thus, VEGF may predispose the lung to inflammation and/or repair by activating local DC. It regulates lung mDC expression of innate immunity effector molecules. The data presented here demonstrate how lung VEGF expression functionally affects local mDC for the transition from the innate response to a Th2-type inflammatory response.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Lung/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Antigens/immunology , Antigens/metabolism , Antigens, CD/metabolism , CD11c Antigen/metabolism , Cathepsins/genetics , Cell Movement/immunology , Chemokines/genetics , Cytokines/genetics , Dendritic Cells/metabolism , Gene Expression/genetics , Gene Expression Regulation/immunology , Lectins, C-Type/metabolism , Lung/cytology , Lung/immunology , Lymph Nodes/cytology , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens , Ovalbumin/immunology , Ovalbumin/metabolism , Receptors, Cell Surface/metabolism , Receptors, Chemokine/genetics , Receptors, Vascular Endothelial Growth Factor/genetics , Vascular Endothelial Growth Factor A/genetics
20.
Am J Respir Cell Mol Biol ; 38(3): 346-53, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17932374

ABSTRACT

Transforming growth factor (TGF)-beta(1) is an essential regulatory cytokine that has been implicated in the pathogenesis of diverse facets of the injury and repair responses in the lung. The types of responses that it elicits can be appreciated in studies from our laboratory that demonstrated that the transgenic (Tg) overexpression of TGF-beta(1) in the murine lung causes epithelial apoptosis followed by fibrosis, inflammation, and parenchymal destruction. Because a cyclin-dependent kinase inhibitor, p21, is a key regulator of apoptosis, we hypothesized that p21 plays an important role in the pathogenesis of TGF-beta(1)-induced tissue responses. To test this hypothesis we evaluated the effect of TGF-beta(1) on the expression of p21 in the murine lung. We also characterized the effects of transgenic TGF-beta(1) in mice with wild-type and null mutant p21 loci. These studies demonstrate that TGF-beta(1) is a potent stimulator of p21 expression in the epithelial cells and macrophages in the murine lung. They also demonstrate that TGF-beta(1)-induced lung inflammation, fibrosis, myofibroblast accumulation, and alveolar destruction are augmented in the absence of p21, and that these alterations are associated with exaggerated levels of apoptosis and caspase-3 activation. Finally, our studies further demonstrated that TGF-beta(1) induces p21 via a TNF-alpha-signaling pathway and that p21 is a negative modulator of TGF-beta(1)-induced TNF-alpha expression. Collectively, our studies demonstrate that p21 regulates TGF-beta(1)-induced apoptosis, inflammation, fibrosis, and alveolar remodeling by interacting with TNF-alpha-signaling pathways.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , Lung/pathology , Signal Transduction , Transforming Growth Factor beta1/toxicity , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/drug effects , Crosses, Genetic , Cyclin-Dependent Kinase Inhibitor p21/genetics , Doxycycline/administration & dosage , Fibrosis/chemically induced , Immunohistochemistry , Inflammation/chemically induced , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/analysis , Random Allocation , Statistics as Topic , Transforming Growth Factor beta1/analysis , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...