Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 384
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Allergy Clin Immunol ; 154(3): 745-753, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38636606

ABSTRACT

BACKGROUND: IgE-mediated degranulation of mast cells (MCs) provides rapid protection against environmental hazards, including animal venoms. A fraction of tissue-resident MCs intimately associates with blood vessels. These perivascular MCs were reported to extend projections into the vessel lumen and to be the first MCs to acquire intravenously injected IgE, suggesting that IgE loading of MCs depends on their vascular association. OBJECTIVE: We sought to elucidate the molecular basis of the MC-blood vessel interaction and to determine its relevance for IgE-mediated immune responses. METHODS: We selectively inactivated the Itgb1 gene, encoding the ß1 chain of integrin adhesion molecules (ITGB1), in MCs by conditional gene targeting in mice. We analyzed skin MCs for blood vessel association, surface IgE density, and capability to bind circulating antibody specific for MC surface molecules, as well as in vivo responses to antigen administered via different routes. RESULTS: Lack of ITGB1 expression severely compromised MC-blood vessel association. ITGB1-deficient MCs showed normal densities of surface IgE but reduced binding of intravenously injected antibodies. While their capacity to degranulate in response to IgE ligation in vivo was unimpaired, anaphylactic responses to antigen circulating in the vasculature were largely abolished. CONCLUSIONS: ITGB1-mediated association of MCs with blood vessels is key for MC immune surveillance of blood vessel content, but is dispensable for slow steady-state loading of endogenous IgE onto tissue-resident MCs.


Subject(s)
Immunoglobulin E , Integrin beta1 , Mast Cells , Animals , Mast Cells/immunology , Integrin beta1/immunology , Integrin beta1/metabolism , Integrin beta1/genetics , Immunoglobulin E/immunology , Mice , Blood Vessels/immunology , Mice, Knockout , Anaphylaxis/immunology , Cell Degranulation/immunology , Mice, Inbred C57BL , Skin/immunology , Skin/blood supply
2.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32817220

ABSTRACT

Lassa fever (LF) is a zoonotic viral hemorrhagic fever caused by Lassa virus (LASV), which is endemic to West African countries. Previous studies have suggested an important role for T-cell-mediated immunopathology in LF pathogenesis, but the mechanisms by which T cells influence disease severity and outcome are not well understood. Here, we present a multiparametric analysis of clinical immunology data collected during the 2017-2018 Lassa fever outbreak in Nigeria. During the acute phase of LF, we observed robust activation of the polyclonal T-cell repertoire, which included LASV-specific and antigenically unrelated T cells. However, severe and fatal LF cases were characterized by poor LASV-specific effector T-cell responses. Severe LF was also characterized by the presence of circulating T cells with homing capacity to inflamed tissues, including the gut mucosa. These findings in LF patients were recapitulated in a mouse model of LASV infection, in which mucosal exposure resulted in remarkably high lethality compared to skin exposure. Taken together, our findings indicate that poor LASV-specific T-cell responses and activation of nonspecific T cells with homing capacity to inflamed tissues are associated with severe LF.IMPORTANCE Lassa fever may cause severe disease in humans, in particular in areas of endemicity like Sierra Leone and Nigeria. Despite its public health importance, the pathophysiology of Lassa fever in humans is poorly understood. Here, we present clinical immunology data obtained in the field during the 2018 Lassa fever outbreak in Nigeria indicating that severe Lassa fever is associated with activation of T cells antigenically unrelated to Lassa virus and poor Lassa virus-specific effector T-cell responses. Mechanistically, we show that these bystander T cells express defined tissue homing signatures that suggest their recruitment to inflamed tissues and a putative role of these T cells in immunopathology. These findings open a window of opportunity to consider T-cell targeting as a potential postexposure therapeutic strategy against severe Lassa fever, a hypothesis that could be tested in relevant animal models, such as nonhuman primates.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Outbreaks , Intestinal Mucosa/immunology , Lassa Fever/immunology , Lassa virus/pathogenicity , Lymphocyte Activation , Adolescent , Adult , Aged , Animals , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Child , Child, Preschool , Female , Gene Expression Regulation , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Infant , Infant, Newborn , Integrin beta1/genetics , Integrin beta1/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Lassa Fever/genetics , Lassa Fever/mortality , Lassa Fever/virology , Lassa virus/growth & development , Lassa virus/immunology , Lysosomal-Associated Membrane Protein 1/genetics , Lysosomal-Associated Membrane Protein 1/immunology , Male , Mice , Middle Aged , Nigeria/epidemiology , Retrospective Studies , Severity of Illness Index , Skin/immunology , Skin/pathology , Skin/virology , Survival Analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
3.
J Immunol ; 202(9): 2782-2794, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30894426

ABSTRACT

Abs exert several of their effector functions by binding to cell surface receptors. For murine IgG3 (mIgG3), the identity of its receptors (and the very existence of a receptor) is still under debate, as not all mIgG3 functions can be explained by interaction with FcγRI. This implies the existence of an alternate receptor, whose identity we sought to pinpoint. We found that blockage of integrin ß1 selectively hampered binding of mIgG3 to macrophages and mIgG3-mediated phagocytosis. Manganese, an integrin activator, increased mIgG3 binding to macrophages. Blockage of FcγRI or Itgb1 inhibited binding of different mIgG3 Abs to variable extents. Our results are consistent with the notion that Itgb1 functions as part of an IgG receptor complex. Given the more ancient origin of integrins in comparison with FcγR, this observation could have far-ranging implications for our understanding of the evolution of Ab-mediated immunity as well as in immunity to microorganisms, pathogenesis of autoimmune diseases, and Ab engineering.


Subject(s)
Immunoglobulin G/immunology , Integrin beta1/immunology , Macrophages/immunology , Phagocytosis , Receptors, IgG/immunology , Animals , Immunoglobulin G/genetics , Integrin beta1/genetics , Mice , Mice, Knockout , Receptors, IgG/genetics
4.
Int J Mol Sci ; 22(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070747

ABSTRACT

Colorectal cancer (CRC) is one of the main causes of cancer death in the world. Post-translational modifications (PTMs) have been extensively studied in malignancies due to its relevance in tumor pathogenesis and therapy. This review is focused on the dysregulation of glycosyltransferase expression in CRC and its impact in cell function and in several biological pathways associated with CRC pathogenesis, prognosis and therapeutic approaches. Glycan structures act as interface molecules between cells and their environment and in several cases facilitate molecule function. CRC tissue shows alterations in glycan structures decorating molecules, such as annexin-1, mucins, heat shock protein 90 (Hsp90), ß1 integrin, carcinoembryonic antigen (CEA), epidermal growth factor receptor (EGFR), insulin-like growth factor-binding protein 3 (IGFBP3), transforming growth factor beta (TGF-ß) receptors, Fas (CD95), PD-L1, decorin, sorbin and SH3 domain-containing protein 1 (SORBS1), CD147 and glycosphingolipids. All of these are described as key molecules in oncogenesis and metastasis. Therefore, glycosylation in CRC can affect cell migration, cell-cell adhesion, actin polymerization, mitosis, cell membrane repair, apoptosis, cell differentiation, stemness regulation, intestinal mucosal barrier integrity, immune system regulation, T cell polarization and gut microbiota composition; all such functions are associated with the prognosis and evolution of the disease. According to these findings, multiple strategies have been evaluated to alter oligosaccharide processing and to modify glycoconjugate structures in order to control CRC progression and prevent metastasis. Additionally, immunotherapy approaches have contemplated the use of neo-antigens, generated by altered glycosylation, as targets for tumor-specific T cells or engineered CAR (Chimeric antigen receptors) T cells.


Subject(s)
Colorectal Neoplasms/genetics , Glycosphingolipids/immunology , Glycosyltransferases/genetics , Mucins/genetics , Neoplasm Proteins/genetics , Protein Processing, Post-Translational , Annexin A1/genetics , Annexin A1/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Decorin/genetics , Decorin/immunology , ErbB Receptors/genetics , ErbB Receptors/immunology , Gene Expression Regulation, Neoplastic , Glycosphingolipids/metabolism , Glycosylation , Glycosyltransferases/immunology , Humans , Immunotherapy, Adoptive/methods , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/immunology , Integrin beta1/genetics , Integrin beta1/immunology , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Mucins/immunology , Neoplasm Proteins/immunology , fas Receptor/genetics , fas Receptor/immunology
5.
Eur J Immunol ; 49(11): 2030-2043, 2019 11.
Article in English | MEDLINE | ID: mdl-31318439

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are found in the CNS during neuroinflammation and have been reported to exert regulatory functions in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). However, the mechanisms of entry of pDCs into the CNS as well as their phenotype and innate functional properties, once recruited into the CNS, have not been thoroughly examined. Herein, we show that pDCs rapidly accumulate into the brain and spinal cord during the acute phase of EAE, and maintain the expression of numerous phenotypic markers typical of peripheral pDCs. Functionally, CNS-pDCs constitutively expressed IRF7 and were able to rapidly produce type I IFNs and IL-12p40 upon ex vivo TLR-9 stimulation. Using adoptive transfer experiments, we provide evidence that CNS-pDC are recruited from the blood and accumulate into the CNS during the acute phase of EAE. Accumulation of pDCs into the CNS was strongly inhibited in the absence of CD29, but not CD18, suggesting a major role for ß1 but not ß2 integrins. Indeed, blocking the CD49d α4-integrins during acute EAE drastically diminished CNS-pDC numbers. Together, our results demonstrate that circulating pDCs are actively recruited into the CNS during acute EAE through a mechanism largely dependent on CD49d/CD29-integrins.


Subject(s)
Brain/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4/immunology , Integrin beta1/immunology , Spinal Cord/immunology , Adoptive Transfer , Animals , Brain/drug effects , Brain/pathology , Cell Movement/immunology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Dendritic Cells/transplantation , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation , Immunity, Innate , Integrin alpha4/genetics , Integrin beta1/genetics , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/genetics , Interferon Type I/immunology , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Peptide Fragments/administration & dosage , Pertussis Toxin/administration & dosage , Signal Transduction , Spinal Cord/drug effects , Spinal Cord/pathology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
6.
Cancer Invest ; 38(4): 228-239, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32208057

ABSTRACT

The aim of this study was to characterize both by flow cytometry analysis and immunohistochemistry cervix uteri cells of nulliparous women screened for cervical intraepithelial neoplasia (CIN) in comparison to a group without CIN by using mesenchymal stem cell-like and hematopoietic lineage markers. A significant expression for CD29, CD38, HLA-I, and HLA-II was correlated positively to the CIN degree and it was more relevant in patients positive for human papilloma virus (HPV). Thus, identification and detailed characterization of pluripotent resident in uteri cells could be a promising therapeutic target.


Subject(s)
Cervix Uteri/cytology , Neoplastic Stem Cells/pathology , Papillomavirus Infections/pathology , Uterine Cervical Dysplasia/pathology , Uterine Cervical Neoplasms/pathology , ADP-ribosyl Cyclase 1/analysis , ADP-ribosyl Cyclase 1/immunology , ADP-ribosyl Cyclase 1/metabolism , Adult , Biopsy , Cervix Uteri/immunology , Cervix Uteri/pathology , Cervix Uteri/virology , Cyclin-Dependent Kinase Inhibitor p16/analysis , Cyclin-Dependent Kinase Inhibitor p16/immunology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Flow Cytometry , Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/analysis , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Immunohistochemistry , Immunophenotyping , Integrin beta1/analysis , Integrin beta1/immunology , Integrin beta1/metabolism , Membrane Glycoproteins/analysis , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Neoplasm Grading , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/virology , Papillomaviridae/immunology , Papillomaviridae/isolation & purification , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Prospective Studies , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Uterine Cervical Dysplasia/immunology , Uterine Cervical Dysplasia/virology
7.
J Hepatol ; 71(6): 1193-1205, 2019 12.
Article in English | MEDLINE | ID: mdl-31433301

ABSTRACT

BACKGROUND & AIMS: Hepatic recruitment of monocyte-derived macrophages (MoMFs) contributes to the inflammatory response in non-alcoholic steatohepatitis (NASH). However, how hepatocyte lipotoxicity promotes MoMF inflammation is unclear. Here we demonstrate that lipotoxic hepatocyte-derived extracellular vesicles (LPC-EVs) are enriched with active integrin ß1 (ITGß1), which promotes monocyte adhesion and liver inflammation in murine NASH. METHODS: Hepatocytes were treated with either vehicle or the toxic lipid mediator lysophosphatidylcholine (LPC); EVs were isolated from the conditioned media and subjected to proteomic analysis. C57BL/6J mice were fed a diet rich in fat, fructose, and cholesterol (FFC) to induce NASH. Mice were treated with anti-ITGß1 neutralizing antibody (ITGß1Ab) or control IgG isotype. RESULTS: Ingenuity® Pathway Analysis of the LPC-EV proteome indicated that ITG signaling is an overrepresented canonical pathway. Immunogold electron microscopy and nanoscale flow cytometry confirmed that LPC-EVs were enriched with activated ITGß1. Furthermore, we showed that LPC treatment in hepatocytes activates ITGß1 and mediates its endocytic trafficking and sorting into EVs. LPC-EVs enhanced monocyte adhesion to liver sinusoidal cells, as observed by shear stress adhesion assay. This adhesion was attenuated in the presence of ITGß1Ab. FFC-fed, ITGß1Ab-treated mice displayed reduced inflammation, defined by decreased hepatic infiltration and activation of proinflammatory MoMFs, as assessed by immunohistochemistry, mRNA expression, and flow cytometry. Likewise, mass cytometry by time-of-flight on intrahepatic leukocytes showed that ITGß1Ab reduced levels of infiltrating proinflammatory monocytes. Furthermore, ITGß1Ab treatment significantly ameliorated liver injury and fibrosis. CONCLUSIONS: Lipotoxic EVs mediate monocyte adhesion to LSECs mainly through an ITGß1-dependent mechanism. ITGß1Ab ameliorates diet-induced NASH in mice by reducing MoMF-driven inflammation, suggesting that blocking ITGß1 is a potential anti-inflammatory therapeutic strategy in human NASH. LAY SUMMARY: Herein, we report that a cell adhesion molecule termed integrin ß1 (ITGß1) plays a key role in the progression of non-alcoholic steatohepatitis (NASH). ITGß1 is released from hepatocytes under lipotoxic stress as a cargo of extracellular vesicles, and mediates monocyte adhesion to liver sinusoidal endothelial cells, which is an essential step in hepatic inflammation. In a mouse model of NASH, blocking ITGß1 reduces liver inflammation, injury and fibrosis. Hence, ITGß1 inhibition may serve as a new therapeutic strategy for NASH.


Subject(s)
Antibodies, Neutralizing , Cell Adhesion/immunology , Hepatocytes/immunology , Integrin beta1/immunology , Lysophosphatidylcholines/pharmacology , Macrophages/immunology , Non-alcoholic Fatty Liver Disease/immunology , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , Diet, High-Fat , Disease Models, Animal , Extracellular Vesicles/immunology , Hepatocytes/metabolism , Humans , Liver Cirrhosis/prevention & control , Mice , Monocytes/immunology , Non-alcoholic Fatty Liver Disease/therapy
8.
J Immunol ; 199(4): 1453-1464, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28674181

ABSTRACT

Robust lung inflammation is one of the prominent features in the pathogenesis of acute lung injury (ALI). Macrophage migration and recruitment are often seen at the early stage of lung inflammatory responses to noxious stimuli. Using an acid inhalation-induced lung injury model, we explored the mechanisms by which acid exposure initiates macrophage recruitment and migration during development of ALI. The lung epithelium comprises a large surface area and functions as a first-line defense against noxious insults. We found that acid exposure induced a remarkable microvesicle (MV) release from lung epithelium as detected in bronchoalveolar lavage fluid. Significantly elevated RNA, rather than protein, was found in these epithelium-derived MVs after acid and included several highly elevated microRNAs, including microRNA (miR)-17 and miR-221. Acid-induced epithelial MV release promoted macrophage migration in vitro and recruitment into the lung in vivo and required, in part, MV shuttling of miR-17 and/or miR-221. Mechanistically, acid-induced epithelial MV miR-17/221 promoted ß1 integrin recycling and presentation back onto the surface of macrophages, in part via a Rab11-mediated pathway. Integrin ß1 is known to play an essential role in regulating macrophage migration. Taken together, acid-induced ALI results in epithelial MV shuttling of miR-17/221 that in turn modulates macrophage ß1 integrin recycling, promoting macrophage recruitment and ultimately contributing to lung inflammation.


Subject(s)
Acute Lung Injury/immunology , Cell-Derived Microparticles , Integrin beta1/metabolism , Lung/cytology , Macrophages/physiology , MicroRNAs/metabolism , Acute Lung Injury/chemically induced , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Movement , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/physiology , Hydrochloric Acid/administration & dosage , Inflammation/immunology , Integrin beta1/immunology , Lung/pathology , Lung/physiology , Macrophages/immunology , Mice , MicroRNAs/isolation & purification
9.
Bull Exp Biol Med ; 167(5): 653-655, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31641985

ABSTRACT

Expression of CD11, CD29, CD36, and DC-STAMP molecules by macrophages was analyzed in in vitro experiments. These molecules mediate cell fusion, one of the mechanisms underlying the formation of multinuclear macrophages. Macrophages were obtained from intact and BCG-infected male BALB/c mice. In intact cultures, multinuclear macrophages appeared primarily due to amitotic division of cell nuclei, while in macrophage cultures from infected mice, the process of cell fusion predominated. In intact macrophage cultures, bi- and multinuclear cells expressed primarily CD29 and CD36. In cultures from infected mice, macrophages expressing CD29 and DC-STAMP predominated, but bi- and multinuclear macrophages expressing CD11 and CD36 predominated over mononuclear ones. The study of macrophage fusion mechanism can be useful for understanding of this biological phenomenon as the mechanisms of delivery of M. tuberculosis and lysosomotropic anti-tuberculosis drugs into tuberculous granulomas to suppress M. tuberculosis persisting in macrophages and reduce the destructive potential of granulomas.


Subject(s)
BCG Vaccine/administration & dosage , CD11 Antigens/genetics , CD36 Antigens/genetics , Integrin beta1/genetics , Macrophages/microbiology , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Animals , CD11 Antigens/immunology , CD36 Antigens/immunology , Cell Fusion , Cell Nucleus/ultrastructure , Gene Expression , Integrin beta1/immunology , Macrophages/immunology , Macrophages/pathology , Male , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/immunology , Primary Cell Culture
10.
Cell Physiol Biochem ; 51(4): 1815-1829, 2018.
Article in English | MEDLINE | ID: mdl-30504725

ABSTRACT

BACKGROUND/AIMS: Mycobacteria-induced diseases, especially tuberculosis, cause more than 1 million deaths each year, which is higher than any other single bacterial pathogen. Neutral sphingomyelinase 2 (Nsm2) has been implied in many physiological processes and diseases, but the role of Nsm2 in pathogen-host interactions and mycobacterial infections has barely been studied. METHODS: We investigated the role of the Nsm2/ceramide system in systemic infection of mice and murine macrophages with Mycobacterium bovis Bacillus Calmette-Guérin (BCG) as a model for mycobacterial infection. For in vitro assays we isolated bone marrow-derived macrophages from Wildtype mice or Nsm2-heterozygous and investigated the role of Nsm2 for macrophage migration/clustering as well as the involvement of p38 mitogen-activated protein kinases (p38K), c-Jun N-terminal kinase (JNK), ß1-integrin and Rac1 activity by Western blot and microscopic studies. For in vivo assays we injected mice intravenously with BCG and analyzed infected tissues for the role of Nsm2-mediated activation of ß1-integrin in granuloma formation and bacterial burden. RESULTS: Our results reveal that BCG infection of macrophages results in rapid stimulation of Nsm2. Genetic and pharmacological studies demonstrate that Nsm2 stimulates a signaling cascade via p38K and JNK to an activation of surface ß1-integrin and Rac1 that leads to the formation of granuloma-like macrophages clusters in vitro and granuloma in vivo. Heterozygosity of Nsm2 in macrophages or antibody-mediated neutralization of active b1-integrin reduced macrophage clusters in vitro and granuloma formation in vivo. Most importantly, Nsm2 heterozygosity or treatment with neutralizing antibodies against ß1-integrin protected mice from systemic BCG infections and chronic infections of the liver and spleen. CONCLUSION: The findings indicate that the Nsm2/ ceramide system plays an important role in systemic infection of mice with mycobacteria by regulating a signaling cascade via p38K, JNK, b1-integrin and Rac1.


Subject(s)
Integrin beta1/immunology , Mycobacterium bovis/immunology , Signal Transduction , Sphingomyelin Phosphodiesterase/immunology , Tuberculosis/veterinary , Animals , Ceramides/immunology , Granuloma/immunology , Granuloma/microbiology , Granuloma/pathology , Granuloma/veterinary , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology
11.
Cell Physiol Biochem ; 50(3): 1015-1028, 2018.
Article in English | MEDLINE | ID: mdl-30355914

ABSTRACT

BACKGROUND/AIMS: The mitogenic effects of periodic mechanical stress on chondrocytes have been studied extensively, but the mechanisms whereby chondrocytes sense and respond to mechanical stimuli remain to be determined. We explored the question and verified the key role of G protein coupled receptor kinase interacting protein 1 (GIT1) signaling in periodic mechanical stress-induced chondrocyte proliferation. METHODS: Two steps were undertaken in the experiment. In the first step, the cells were maintained under non-pressure conditions or periodic mechanical stress for 1 h prior to Western blot analysis. In the second step, the cells were pretreated with short hairpin RNA (shRNA) targeted to GIT1 or Src or control scrambled shRNA, or transfected with GIT1 wild-type or GIT1 mutant Y321F, or focal adhesion kinase (FAK) wild-type or FAK mutants Y397F or Y576F/Y577, respectively. Moreover, the cells were pretreated with blocking antibody against integrin ß1 or PP2. Then the cells were maintained under non-pressure conditions or periodic mechanical stress for 1 h prior to Western blot analysis, and for 3 days, 8 h per day, prior to direct cell counting and CCK-8 assay, respectively. RESULTS: Periodic mechanical stress significantly induced sustained phosphorylation of GIT1 at Tyr321. Reduction of GIT1 with shRNA targeted to GIT1 and GIT1 mutant Y321F inhibited periodic mechanical stress-promoted chondrocyte proliferation, accompanied by attenuated extracellular signal-regulated kinase (ERK)1/2 and FAK phosphorylation at Tyr576/577. However, activation of Src and FAK-Tyr397 was not prevented upon GIT1 suppression. Furthermore, pretreatment with blocking antibody against integrin ß1, Src-selective inhibitor, PP2, and shRNA targeted to Src blocked GIT1 activation under periodic mechanical stress. In addition, GIT1 phosphorylation at Tyr321 was not reduced upon pretreatment with FAK mutants Y397F or Y576F/Y577 under conditions of periodic mechanical stress. CONCLUSION: These findings collectively suggested that periodic mechanical stress promoted chondrocyte proliferation through at least two separate pathways, integrin ß1-Src-GIT1-FAK(Tyr576/577)-ERK1/2, and the other parallel GIT1-independent integrin ß1-FAK(Tyr397)-ERK1/2.


Subject(s)
Cell Cycle Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphoproteins/metabolism , Stress, Mechanical , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Integrin beta1/immunology , Integrin beta1/metabolism , Mutagenesis, Site-Directed , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , src-Family Kinases/metabolism
12.
Immunology ; 152(2): 308-327, 2017 10.
Article in English | MEDLINE | ID: mdl-28580688

ABSTRACT

Cell adhesion is generally considered to depend on positive regulation through ligation of integrins and cytokine receptors. However, here we show that T-cell adhesion, and notably also T-cell receptor (TCR) -induced activation, are subject to constant suppression through shedding of low-density lipoprotein receptor-related protein 1 (LRP1). The broad-spectrum metalloprotease inhibitor GM6001 abrogated shedding, so inducing prominent cell surface expression of LRP1 while enhancing TCR-induced activation and adhesion to ß1 and ß2 integrin ligands, hence arresting the cells. Integrin ligands also inhibited shedding but the effect was less potent than that of GM6001. Unlike GM6001, integrin ligands also induced cell surface expression of full-length thrombospondin-1 (TSP170) and TSP130, which associated with LRP1, and TSP110, which did not associate with LRP1. Cell surface expression of LRP1 and TSP130 were induced exclusively in adhering cells, expression of TSP110 preferentially in non-adhering cells and expression of TSP170 correlated with T-cell motility. The pro-adhesive chemokine CXCL12 also inhibited LRP1 shedding and induced surface expression of TSP170 and TSP130 while inhibiting TSP110. Exogenous TSP-1 and ligation of CD28 inhibited shedding although less effectively than GM6001, and the inhibition through CD28 was independent of TSP-1. Small interfering RNA silencing experiments confirmed involvement of LRP1 and TSP-1 in integrin-dependent adhesion and TCR-induced activation. Hence, the poor LRP1 expression in T cells depends on shedding. Integrin ligands and CXCL12 antagonize shedding through a TSP-1-dependent pathway and ligation of CD28 antagonizes shedding independent of TSP-1. The disappearance of LRP1 from the cell surface may provide basic immunosuppression at the T-cell level.


Subject(s)
Cell Adhesion , Cell Movement , Immune Tolerance , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Lymphocyte Activation , T-Lymphocytes/metabolism , CD18 Antigens/immunology , CD18 Antigens/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Chemokine CXCL12/pharmacology , Dipeptides/pharmacology , Humans , Immune Tolerance/drug effects , Integrin beta1/immunology , Integrin beta1/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/immunology , Lymphocyte Activation/drug effects , Matrix Metalloproteinase Inhibitors/pharmacology , Peptide Fragments/immunology , Peptide Fragments/metabolism , RNA Interference , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Thrombospondin 1/genetics , Thrombospondin 1/immunology , Thrombospondin 1/metabolism , Time Factors , Transfection
13.
Biochem Biophys Res Commun ; 491(3): 629-635, 2017 09 23.
Article in English | MEDLINE | ID: mdl-28760342

ABSTRACT

Acupuncture therapy is performed by applying the needle insertion at discrete cutaneous locations and used for the treatments of diverse symptoms and disorders. In order to elucidate mechanistic basis on how acupuncture stimulation (AS) produces therapeutic effects, it is primarily important to understand tissue responses locally at the acupuncture site (acupoint). Here, we investigated integrin protein as molecular target responding to and integrating AS. Signals of α6 and ß1 integrins were clearly induced at zusanli acupoint 24 h after AS in areas of nuclear clusters around the needle track. Induction levels of integrin were largely reduced by needle insertion at non-acupuncture point or without needle rotation. Phospho-Erk1/2 was initially decreased below the basal level after AS but increased 24 h later. Induction pattern of phospho-Erk1/2 was as similar as that of α6 integrin in its selectivity to needling procedure and tissue distribution. We further found that mRNA expression of P2X3 purinergic receptor was upregulated in the dorsal root ganglion (DRG) after AS, but decreased by the inhibition of Erk1/2 activity at the acupuncture area. Moreover, AS-mediated integrin activation was required for Erk1/2 activation at the acupuncture site and regulation of pain sensitivity in the hind paw. The present results provide a new evidence on acupuncture-specific tissue response in terms of integrin induction, and further suggest that integrin activation may be involved in transmitting mechanosensory signals from the acupoint to afferent nerve fiber.


Subject(s)
Acupuncture Therapy/methods , Integrin alpha6/immunology , Integrin beta1/immunology , MAP Kinase Signaling System/immunology , Neuralgia/immunology , Neuralgia/therapy , Acupuncture Points , Animals , Male , Mechanotransduction, Cellular/immunology , Neuralgia/diagnosis , Physical Stimulation/methods , Rats , Rats, Sprague-Dawley , Treatment Outcome
14.
Clin Exp Immunol ; 190(3): 315-327, 2017 12.
Article in English | MEDLINE | ID: mdl-28862740

ABSTRACT

The role of macrophage infiltrates in oral mucosal acute graft-versus-host disease (AGVHD) remains unclear, although clinical studies suggest that macrophage infiltration correlates directly with the severity of AGVHD. In this study, we investigated the role of M1 macrophage infiltration in the oral mucosa of rats with AGVHD. Lewis rat spleen cells were injected into (Lewis × Brown Norway) F1 rats to induce systemic GVHD. Tongue samples were evaluated using histology, immunohistochemistry, dual immunofluorescence, real-time reverse transcription-polymerase chain reaction, Transwell migration assays and Stamper-Woodruff binding assays. At the onset of oral mucosal AGVHD, dual immunofluorescence and migration assays revealed that M1 macrophages had accumulated in the basement membrane (BM) region via the laminin/CD29 ß1 integrin pathway. Macrophage-secreted matrix metalloproteinase-2 was related to BM degradation. The adhesion of macrophages to the oral epithelium could be inhibited by pretreating macrophages with a CC chemokine receptor 2 (CCR2) antibody and/or pretreating lesion sections with monocyte chemoattractant protein-1 (MCP-1) antibody. Our data show that the migration and adhesion of M1 macrophages are associated with oral mucosal AGVHD, which is mediated in part by both laminin/CD29 ß 1 intern and MCP-1/CCR2 pathways. Therefore, our study provides additional support for the contribution of macrophage infiltrate to the development of oral mucosal AGVHD.


Subject(s)
Cell Movement/immunology , Graft vs Host Disease/immunology , Macrophages/microbiology , Mouth Mucosa/immunology , Acute Disease , Animals , Chemokine CCL2/immunology , Female , Graft vs Host Disease/pathology , Integrin beta1/immunology , Laminin/immunology , Macrophages/pathology , Male , Matrix Metalloproteinase 2/immunology , Mouth Mucosa/pathology , Rats , Rats, Inbred Lew , Receptors, CCR2/immunology
15.
Eur J Immunol ; 44(4): 1156-69, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24338844

ABSTRACT

Integrin trafficking, including internalization, recycling, and lysosomal degradation, is crucial for the regulation of cellular functions. Exosomes, nano-sized extracellular vesicles, are believed to play important roles in intercellular communications. This study demonstrates that exosomes released from human macrophages negatively regulate endothelial cell migration through control of integrin trafficking. Macrophage-derived exosomes promote internalization of integrin ß1 in primary HUVECs. The internalized integrin ß1 persistently accumulates in the perinuclear region and is not recycled back to the plasma membrane. Experimental results indicate that macrophage-derived exosomes stimulate trafficking of internalized integrin ß1 to lysosomal compartments with a corresponding decrease in the integrin destined for recycling endosomes, resulting in proteolytic degradation of the integrin. Moreover, ubiquitination of HUVEC integrin ß1 is enhanced by the exosomes, and exosome-mediated integrin degradation is blocked by bafilomycin A, a lysosomal degradation inhibitor. Macrophage-derived exosomes were also shown to effectively suppress collagen-induced activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase signaling pathway and HUVEC migration, which are both dependent on integrin ß1. These observations provide new insight into the functional significance of exosomes in the regulation of integrin trafficking.


Subject(s)
Cell Movement/immunology , Exosomes/immunology , Human Umbilical Vein Endothelial Cells/immunology , Integrin beta1/immunology , Macrophages/immunology , Cell Line, Tumor , Cells, Cultured , Down-Regulation/immunology , Endocytosis/immunology , Exosomes/ultrastructure , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunoblotting , Integrin beta1/genetics , Integrin beta1/metabolism , Lysosomes/immunology , Lysosomes/metabolism , MAP Kinase Signaling System/immunology , Macrophages/metabolism , Microscopy, Electron, Scanning , Mitogen-Activated Protein Kinases/immunology , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/immunology , Protein Transport/immunology , RNA Interference
16.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L463-75, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24337923

ABSTRACT

Creation of bioartificial organs has been enhanced by the development of strategies involving decellularized mammalian lung. Because fibroblasts critically support lung function through a number of mechanisms, study of these cells in the context of the decellularized lung has the potential to improve the structure and function of tissue-engineered lungs. We characterized the engraftment and survival of a mouse fibroblast cell line in decellularized rat lung slices and found a time-dependent increase in cell numbers assessed by hematoxylin and eosin staining, cell proliferation assessed by Ki67 staining, and minimal cell death assessed by TUNEL staining. We developed a repopulation index to allow quantification of cell survival that accounts for variation in cell density throughout the seeded scaffold. We then applied this method to the study of mouse lung scaffolds and found that decellularization of presliced mouse lungs produced matrices with preserved alveolar architecture and proteinaceous components including fibronectin, collagens I and IV, laminin, and elastin. Treatment with a ß1-integrin-neutralizing antibody significantly reduced the repopulation index after 24 h of culture. Treatment with focal adhesion kinase (FAK) inhibitor and extracellular signal-regulated kinase (ERK) inhibitor further reduced initial repopulation scores while treatment with AKT inhibitor increased initial scores. Rho-associated kinase inhibitor had no discernible effect. These data indicate that initial adhesion and survival of mouse fibroblasts in the decellularized mouse lung occur in a ß1-integrin-dependent, FAK/ERK-dependent manner that is opposed by AKT.


Subject(s)
Bioartificial Organs , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/transplantation , Focal Adhesion Kinase 1/metabolism , Integrin beta1/metabolism , Lung/physiology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antibodies, Neutralizing/immunology , Cell Adhesion/physiology , Cell Line , Cell Proliferation , Cell Survival , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibroblasts/physiology , Focal Adhesion Kinase 1/antagonists & inhibitors , Integrin beta1/immunology , Lung/cytology , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats , Tissue Engineering/methods , Tissue Scaffolds , rho-Associated Kinases/antagonists & inhibitors
17.
Clin Exp Immunol ; 176(1): 120-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24329680

ABSTRACT

Extracorporeal photopheresis (ECP) has been used as a prophylactic and therapeutic option to avoid and treat rejection after heart transplantation (HTx). Tolerance-inducing effects of ECP such as up-regulation of regulatory T cells (T(regs)) are known, but specific effects of ECP on regulatory T cell (T(reg)) subsets and dendritic cells (DCs) are lacking. We analysed different subsets of T(regs) and DCs as well as the immune balance status during ECP treatment after HTx. Blood samples were collected from HTx patients treated with ECP for prophylaxis (n = 9) or from patients with histologically proven acute cellular rejection (ACR) of grade ≥ 1B (n = 9), as well as from control HTx patients without ECP (HTxC; n = 7). Subsets of T(regs) and DCs as well as different cytokine levels were analysed. Almost 80% of the HTx patients showed an effect to ECP treatment with an increase of T(regs) and plasmacytoid DCs (pDCs). The percentage of pDCs before ECP treatment was significantly higher in patients with no ECP effect (26·3% ± 5·6%) compared to patients who showed an effect to ECP (9·8% ± 10·2%; P = 0·011). Analysis of functional subsets of CD4⁺CD25(high)CD127(low) T(regs) showed that CD62L-, CD120b- and CD147-positive T(regs) did not differ between the groups. CD39-positive T(regs) increased during ECP treatment compared to HTxC. ECP-treated patients showed higher levels for T helper type 1 (Th1), Th2 and Th17 cytokines. Cytokine levels were higher in HTx patients with rejection before ECP treatment compared to patients with prophylactic ECP treatment. We recommend a monitoring strategy that includes the quantification and analysis of T(regs), pDCs and the immune balance status before and up to 12 months after starting ECP.


Subject(s)
Graft Rejection/immunology , Heart Transplantation/methods , Monitoring, Immunologic/methods , Photopheresis/methods , Acute Disease , Adult , Aged , Basigin/immunology , Basigin/metabolism , CD3 Complex/immunology , CD3 Complex/metabolism , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Female , Graft Rejection/blood , Humans , Integrin beta1/immunology , Integrin beta1/metabolism , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7 Receptor alpha Subunit/immunology , Interleukin-7 Receptor alpha Subunit/metabolism , Male , Middle Aged , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Time Factors
18.
BMC Cell Biol ; 14: 5, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23350814

ABSTRACT

BACKGROUND: The interactions between stem cells and extracellular matrix (ECM) mediated by integrins play important roles in the processes that determine stem cell fate. However, the role of ECM/integrin interaction in the formation of embryoid bodies (EBs) during cardiogenesis from murine induced pluripotent stem cells (miPSCs) remains unclear. RESULTS: In the present study, collagen type I and ß(1) integrin were expressed and upregulated synergistically during the formation of miPSC-derived EBs, with a peak expression at day 3 of differentiation. The blockage of collagen/ß(1) integrin interaction by ß(1) integrin blocking antibody resulted in the production of defective EBs that were characterized by decreased size and the absence of a shell-like layer composed of primitive endoderm cells. The quantification of spontaneous beating activity, cardiac-specific gene expression and cardiac troponin T (cTnT) immunostaining showed that the cardiac differentiation of these defective miPSC-derived EBs was lower than that of control EBs. CONCLUSIONS: These findings indicate that collagen/ß(1) integrin interaction is required for the growth and cardiac differentiation of miPSC-derived EBs and will be helpful in future engineering of the matrix microenvironment within EBs to efficiently direct the cardiac fate of pluripotent stem cells to promote cardiovascular regeneration.


Subject(s)
Collagen Type I/metabolism , Embryoid Bodies/cytology , Induced Pluripotent Stem Cells/metabolism , Integrin beta1/metabolism , Animals , Antibodies/immunology , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Immunohistochemistry , Induced Pluripotent Stem Cells/cytology , Integrin beta1/immunology , Mice , Microscopy, Electron, Scanning , Myocardium/cytology , Protein Binding , Troponin T/metabolism
19.
Blood ; 118(7): 1818-27, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21677313

ABSTRACT

CD81 (TAPA-1) is a member of the widely expressed and evolutionary conserved tetraspanin family that forms complexes with a variety of other cell surface receptors and facilitates hepatitis C virus entry. Here, we show that CD81 is specifically required for the formation of lamellipodia in migrating dendritic cells (DCs). Mouse CD81(-/-) DCs, or murine and human CD81 RNA interference knockdown DCs lacked the ability to form actin protrusions, thereby impairing their motility dramatically. Moreover, we observed a selective loss of Rac1 activity in the absence of CD81, the latter of which is exclusively required for integrin-dependent migration on 2-dimensional substrates. Neither integrin affinity for substrate nor the size of basal integrin clusters was affected by CD81 deficiency in adherent DCs. However, the use of total internal reflection fluorescence microscopy revealed an accumulation of integrin clusters above the basal layer in CD81 knockdown cells. Furthermore, ß1- or ß2-integrins, actin, and Rac are strongly colocalized at the leading edge of DCs, but the very fronts of these cells protrude CD81-containing membranes that project outward from the actin-integrin area. Taken together, these data suggest a thus far unappreciated role for CD81 in the mobilization of preformed integrin clusters into the leading edge of migratory DCs on 2-dimensional surfaces.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Actins/immunology , Animals , Antigens, CD/genetics , Cell Adhesion , Cell Movement , Cells, Cultured , Gene Knockdown Techniques , Humans , Integrin beta1/immunology , Integrins/immunology , Mice , Pseudopodia/immunology , Tetraspanin 28 , rac1 GTP-Binding Protein/immunology
20.
Allergy ; 68(10): 1259-68, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24112102

ABSTRACT

BACKGROUND: Mast cell (MC) - eosinophil (Eos) activating cross-talk might be critical for the severity and chronicity of allergy. Among soluble mediators, eosinophil major basic protein (MBP), a hallmark of allergy, is particularly important because it was shown to activate specific MC subtypes. We previously demonstrated that MBP activates IgE-desensitized rat MC and human lung and cord blood-derived MC (CBMC) after priming with fibroblast membranal stem cell factor. However, a distinct mechanism for this activation was missing. Therefore, we aimed to investigate it. METHODS: Major basic protein-1 activation of CBMC primed with fibroblast-derived membranes (FBM) was measured by ß-hexosaminidase and tryptase release. Chemical cross-linking followed by micrometric flow cytometry probed direct interactions. Antibodies neutralized integrin-ß1 and recognized its active form. Pertussis toxin (Ptx) was used to decrease integrin-ß1 active form expression. Hematopoietic cell kinase (Hck) was identified by immunoprecipitation (IP) and silenced by siRNA. RESULTS: Major basic protein-1-induced CBMC activation is mediated partly by MBP1-integrin-ß1 interaction on the MC surface. FBM prime CBMC via a G protein, as confirmed by Ptx, to shift integrin-ß1 to its active form. Following MBP1 binding, integrin-ß1 binds Hck that further transduces the activation signal. MC priming with FBM leads to up-regulation in Hck protein level. MC integrin-ß1 neutralization inhibits MBP1-induced activation and uptake. Hck silencing results with reduced MBP1-induced activation. CONCLUSIONS: Fibroblast-derived membranes, integrin-ß1, and Hck are involved in MBP1-induced activation of CBMC and therefore represent a distinct mechanism for this activation. This finding might implicate integrin-ß1 and Hck as targets for decreasing MC - Eos activating cross-talk in allergy.


Subject(s)
Cell Membrane/immunology , Eosinophil Major Basic Protein/immunology , Eosinophils/immunology , Eosinophils/metabolism , Fibroblasts/immunology , Integrin beta1/immunology , Mast Cells/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Cell Communication/immunology , Cell Membrane/metabolism , Eosinophil Major Basic Protein/metabolism , Fibroblasts/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Silencing , Humans , Integrin beta1/metabolism , Mice , Protein Binding , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL