1
|
Qian WJ, Yan JS, Gang XY, Xu L, Shi S, Li X, Na FJ, Cai LT, Li HM, Zhao MF. Intercellular adhesion molecule-1 (ICAM-1): From molecular functions to clinical applications in cancer investigation. Biochim Biophys Acta Rev Cancer 2024; 1879:189187. [PMID: 39317271 DOI: 10.1016/j.bbcan.2024.189187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Intercellular adhesion molecule-1 (ICAM-1) is a versatile molecule that plays a critical role in various physiological and pathological processes, particularly in tumor development where its impact is bidirectional. On the one hand, it augments the immune response by promoting immune cell migration, infiltration, and the formation of immunological synapses, thus facilitating potent antitumor effects. Simultaneously, it contributes to tumor immune evasion and influences metastasis by mediating transendothelial migration (TEM), epithelial-to-mesenchymal transition (EMT), and epigenetic modification of tumor cells. Despite its significant potential, the full clinical utility of ICAM-1 has yet to be fully realized. In this review, we thoroughly examine recent advancements in understanding the role of ICAM-1 in tumor development, its relevance in predicting therapeutic efficacy and prognosis, as well as the progress in clinical translational research on anti-ICAM-1-based therapies, encompassing including monoclonal antibodies, immunotherapy, antibody-drug conjugate (ADC), and conventional treatments. By shedding light on these innovative strategies, we aim to underscore ICAM-1's significance as a valuable and multifaceted target for cancer treatment, igniting enthusiasm for further research and facilitating translation into clinical applications.
Collapse
Affiliation(s)
- Wen-Jing Qian
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jin-Shan Yan
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Xiao-Yu Gang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Lu Xu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Sha Shi
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Xin Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Fang-Jian Na
- Network Information Center, China Medical University, Shenyang, China
| | - Lu-Tong Cai
- Psychological Medicine, Shenyang Medical College, Shenyang, China
| | - He-Ming Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China; Guangdong Association of Clinical Trials (GACT)/Chinese Thoracic Oncology Group (CTONG) and Guangdong Provincial Key Lab of Translational Medicine in Lung Cancer, Guangzhou, China.
| | - Ming-Fang Zhao
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
2
|
Immler R, Nussbaumer K, Doerner A, El Bounkari O, Huber S, Abisch J, Napoli M, Schmidt S, Margraf A, Pruenster M, Rohwedder I, Lange-Sperandio B, Mall MA, de Jong R, Ohnmacht C, Bernhagen J, Voehringer D, Marth JD, Frommhold D, Sperandio M. CCR3-dependent eosinophil recruitment is regulated by sialyltransferase ST3Gal-IV. Proc Natl Acad Sci U S A 2024; 121:e2319057121. [PMID: 38687790 PMCID: PMC11087806 DOI: 10.1073/pnas.2319057121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Eosinophil recruitment is a pathological hallmark of many allergic and helminthic diseases. Here, we investigated chemokine receptor CCR3-induced eosinophil recruitment in sialyltransferase St3gal4-/- mice. We found a marked decrease in eosinophil extravasation into CCL11-stimulated cremaster muscles and into the inflamed peritoneal cavity of St3gal4-/- mice. Ex vivo flow chamber assays uncovered reduced adhesion of St3gal4-/- compared to wild type eosinophils. Using flow cytometry, we show reduced binding of CCL11 to St3gal4-/- eosinophils. Further, we noted reduced binding of CCL11 to its chemokine receptor CCR3 isolated from St3gal4-/- eosinophils. This was accompanied by almost absent CCR3 internalization of CCL11-stimulated St3gal4-/- eosinophils. Applying an ovalbumin-induced allergic airway disease model, we found a dramatic reduction in eosinophil numbers in bronchoalveolar lavage fluid following intratracheal challenge with ovalbumin in St3gal4-deficient mice. Finally, we also investigated tissue-resident eosinophils under homeostatic conditions and found reduced resident eosinophil numbers in the thymus and adipose tissue in the absence of ST3Gal-IV. Taken together, our results demonstrate an important role of ST3Gal-IV in CCR3-induced eosinophil recruitment in vivo rendering this enzyme an attractive target in reducing unwanted eosinophil infiltration in various disorders including allergic diseases.
Collapse
Affiliation(s)
- Roland Immler
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-UniversitätMünchen, PLanegg-Martinsried82152, Germany
| | - Katrin Nussbaumer
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-UniversitätMünchen, PLanegg-Martinsried82152, Germany
| | - Axel Doerner
- Department of Neonatology, University of Heidelberg, Heidelberg69120, Germany
| | - Omar El Bounkari
- Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität, München81377, Germany
| | - Silke Huber
- Institute of Immunology, Ludwig-Maximilians-Universität München, München80336, Germany
| | - Janine Abisch
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-UniversitätMünchen, PLanegg-Martinsried82152, Germany
| | - Matteo Napoli
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-UniversitätMünchen, PLanegg-Martinsried82152, Germany
| | - Sarah Schmidt
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-UniversitätMünchen, PLanegg-Martinsried82152, Germany
| | - Andreas Margraf
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-UniversitätMünchen, PLanegg-Martinsried82152, Germany
| | - Monika Pruenster
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-UniversitätMünchen, PLanegg-Martinsried82152, Germany
| | - Ina Rohwedder
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-UniversitätMünchen, PLanegg-Martinsried82152, Germany
| | - Baerbel Lange-Sperandio
- von Haunersches Kinderspital, Klinikum der Universität München, Ludwig-Maximilians-Universität, München80336, Germany
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin13353, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin10117, Germany
- German Centre for Lung Research, Associated Partner Site, Berlin13353, Germany
| | - Renske de Jong
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, München80802, Germany
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, München80802, Germany
| | - Juergen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität, München81377, Germany
- Munich Cluster for Systems Neurology, München81377, Germany
- Munich Heart Alliance, München80336, Germany
| | - David Voehringer
- Institute of Immunology, Ludwig-Maximilians-Universität München, München80336, Germany
- Department of Infection Biology, University of Erlangen, Erlangen91054, Germany
| | - Jamey D. Marth
- Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Diseases, San Diego, CA92037
| | - David Frommhold
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-UniversitätMünchen, PLanegg-Martinsried82152, Germany
- Children’s Hospital Memmingen, Memmingen87700, Germany
| | - Markus Sperandio
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-UniversitätMünchen, PLanegg-Martinsried82152, Germany
| |
Collapse
|
3
|
Henrioux F, Navel V, Belville C, Charnay C, Antoine A, Chiambaretta F, Sapin V, Blanchon L. Inflammation of Dry Eye Syndrome: A Cellular Study of the Epithelial and Macrophagic Involvement of NFAT5 and RAGE. Int J Mol Sci 2023; 24:11052. [PMID: 37446230 DOI: 10.3390/ijms241311052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/20/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Dry eye inflammation is a key step in a vicious circle and needs to be better understood in order to break it. The goals of this work were to, first, characterize alarmins and cytokines released by ocular surface cells in the hyperosmolar context and, second, study the role of NFAT5 in this process. Finally, we studied the potential action of these alarmins in ocular surface epithelial cells and macrophages via RAGE pathways. HCE and WKD cell lines were cultured in a NaCl-hyperosmolar medium and the expression of alarmins (S100A4, S100A8, S100A9, and HMGB1), cytokines (IL6, IL8, TNFα, and MCP1), and NFAT5 were assessed using RT-qPCR, ELISA and multiplex, Western blot, immunofluorescence, and luciferase assays. In selected experiments, an inhibitor of RAGE (RAP) or NFAT5 siRNAs were added before the hyperosmolar stimulations. HCE and WKD cells or macrophages were treated with recombinant proteins of alarmins (with or without RAP) and analyzed for cytokine expression and chemotaxis, respectively. Hyperosmolarity induced epithelial cell inflammation depending on cell type. NFAT5, but not RAGE or alarmins, participated in triggering epithelial inflammation. Furthermore, the release of alarmins induced macrophage migration through RAGE. These in vitro results suggest that NFAT5 and RAGE have a role in dry eye inflammation.
Collapse
Affiliation(s)
- Fanny Henrioux
- Team "Translational Approach to Epithelial Injury and Repair", Institute Genetics, Reproduction and Development (iGReD), Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Valentin Navel
- Team "Translational Approach to Epithelial Injury and Repair", Institute Genetics, Reproduction and Development (iGReD), Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- Ophthalmology Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Corinne Belville
- Team "Translational Approach to Epithelial Injury and Repair", Institute Genetics, Reproduction and Development (iGReD), Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Coline Charnay
- Team "Translational Approach to Epithelial Injury and Repair", Institute Genetics, Reproduction and Development (iGReD), Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Audrey Antoine
- Team "Translational Approach to Epithelial Injury and Repair", Institute Genetics, Reproduction and Development (iGReD), Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Frédéric Chiambaretta
- Team "Translational Approach to Epithelial Injury and Repair", Institute Genetics, Reproduction and Development (iGReD), Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- Ophthalmology Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Vincent Sapin
- Team "Translational Approach to Epithelial Injury and Repair", Institute Genetics, Reproduction and Development (iGReD), Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- Biochemistry and Molecular Genetics Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Loïc Blanchon
- Team "Translational Approach to Epithelial Injury and Repair", Institute Genetics, Reproduction and Development (iGReD), Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
4
|
Actin Cross-Linking Effector Domain of the Vibrio vulnificus F-Type MARTX Toxin Dominates Disease Progression During Intestinal Infection. Infect Immun 2022; 90:e0062721. [PMID: 35254094 DOI: 10.1128/iai.00627-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Vibrio vulnificus is an opportunistic pathogen that causes gastroenteritis and septicemia in humans. The V. vulnificus multifunctional-autoprocessing repeats-in-toxin (MARTX) toxin is a pore-forming toxin that translocates multiple functionally independent effector domains into target cells and an essential virulence factor for fatal disease. The effector repertoire delivered and thus the mechanism of action of the toxin can differ dramatically across V. vulnificus isolates. Here, we utilize a strain of V. vulnificus that carries an F-type MARTX toxin that delivers an actin cross-linking domain (ACD) and four other effector domains. We demonstrate that ACD is the primary driver of virulence following intragastric infection and of bacterial dissemination to distal organs. We additionally show that ACD activates the transcription of intermediate early response genes in cultured intestinal epithelial cells (IECs). However, the genes activated by ACD are suppressed, at least in part, by the codelivered Ras/Rap1-specific endopeptidase (RRSP). The transcriptional response induced by strains translocating only RRSP results in a unique transcriptional profile, demonstrating that the transcriptional response to V. vulnificus is remodeled rather than simply suppressed by the MARTX toxin effector repertoire. Regardless, the transcriptional response in the intestinal tissue of infected mice is dominated by ACD-mediated induction of genes associated with response to tissue damage and is not impacted by RRSP or the three other effectors codelivered with ACD and RRSP. These data demonstrate that while other effectors do remodel early intestinal innate immune responses, ACD is the dominant driver of disease progression by ACD+ V. vulnificus during intestinal infection.
Collapse
|
5
|
Li H, Li Y, Zhang Y, Tan B, Huang T, Xiong J, Tan X, Ermolaeva MA, Fu L. MAPK10 Expression as a Prognostic Marker of the Immunosuppressive Tumor Microenvironment in Human Hepatocellular Carcinoma. Front Oncol 2021; 11:687371. [PMID: 34408980 PMCID: PMC8366563 DOI: 10.3389/fonc.2021.687371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) remains a devastating malignancy worldwide due to lack of effective therapy. The immune-rich contexture of HCC tumor microenvironment (TME) makes this tumor an appealing target for immune-based therapies; however, the immunosuppressive TME is still a major challenge for more efficient immunotherapy in HCC. Using bioinformatics analysis based on the TCGA database, here we found that MAPK10 is frequently down-regulated in HCC tumors and significantly correlates with poor survival of HCC patients. HCC patients with low MAPK10 expression have lower expression scores of tumor infiltration lymphocytes (TILs) and stromal cells in the TME and increased scores of tumor cells than those with high MAPK10 expression. Further transcriptomic analyses revealed that the immune activity in the TME of HCC was markedly reduced in the low-MAPK10 group of HCC patients compared to the high-MAPK10 group. Additionally, we identified 495 differentially expressed immune-associated genes (DIGs), with 482 genes down-regulated and 13 genes up-regulated in parallel with the decrease of MAPK10 expression. GO enrichment and KEGG pathway analyses indicated that the biological functions of these DIGs included cell chemotaxis, leukocyte migration and positive regulation of the response to cytokine–cytokine receptor interaction, T cell receptor activation and MAPK signaling pathway. Protein–protein interaction (PPI) analyses of the 495 DIGs revealed five potential downstream hub genes of MAPK10, including SYK, CBL, VAV1, LCK, and CD3G. Several hub genes such as SYK, LCK, and VAV1 could respond to the immunological costimulatory signaling mediated by the transmembrane protein ICAM1, which was identified as a down-regulated DIG associated with low-MAPK10 expression. Moreover, ectopic overexpression or knock-down of MAPK10 could up-regulate or down-regulate ICAM1 expression via phosphorylation of c-jun at Ser63 in HCC cell lines, respectively. Collectively, our results demonstrated that MAPK10 down-regulation likely contributes to the immunosuppressive TME of HCC, and this gene might serve as a potential immunotherapeutic target and a prognostic factor for HCC patients.
Collapse
Affiliation(s)
- Huahui Li
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China.,Group of Homeostasis and Stress Tolerance, Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany.,Shenzhen University-Friedrich Schiller Universitat Jena Joint PhD Program in Biomedical Sciences, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuting Li
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China.,Group of Homeostasis and Stress Tolerance, Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany.,Shenzhen University-Friedrich Schiller Universitat Jena Joint PhD Program in Biomedical Sciences, Shenzhen University School of Medicine, Shenzhen, China
| | - Ying Zhang
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Binbin Tan
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Tuxiong Huang
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Jixian Xiong
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Xiangyu Tan
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Maria A Ermolaeva
- Group of Homeostasis and Stress Tolerance, Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Li Fu
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
6
|
Wyczanska M, Lange-Sperandio B. DAMPs in Unilateral Ureteral Obstruction. Front Immunol 2020; 11:581300. [PMID: 33117389 PMCID: PMC7575708 DOI: 10.3389/fimmu.2020.581300] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022] Open
Abstract
Damage-associated molecular patterns (DAMPs) are released from tubular and interstitial cells in the kidney after unilateral ureteral obstruction (UUO). DAMPs are recognized by pattern recognition receptors (PRRs), which mediate the initiation of an immune response and the release of inflammatory cytokines. The animal model of UUO is used for various purposes. UUO in adult mice serves as a model for accelerated renal fibrosis, which is a hallmark of progressive renal disease. UUO in adult mice enables to study cell death, inflammation, and extracellular matrix deposition in the kidney. Neonatal UUO is a model for congenital obstructive nephropathies. It studies inflammation, apoptosis, and interstitial fibrosis in the neonatal kidney, when nephrogenesis is still ongoing. Following UUO, several DAMPs as well as DAMP receptors are upregulated. In adult UUO, soluble uric acid is upregulated and activates the NOD-like receptor family, pyrin domain containing-3 (NLRP3) inflammasome, which promotes fibrosis, apoptosis, and reactive oxygen species (ROS) injury. Further DAMPs associated with UUO are uromodulin, members of the IL-1 family, and necrotic cell DNA, all of which promote sterile inflammation. In neonatal UUO, the receptor for advanced glycation endproducts (RAGE) is highly upregulated. RAGE is a ligand for several DAMPs, including high mobility group box 1 (HMGB1) and S100 proteins, which play an important role in renal fibrosis. Additionally, necroptosis is an important mechanism of cell death, besides apoptosis, in neonatal UUO. It is highly inflammatory due to release of cytokines and specific DAMPs. The release and recognition of DAMPs initiate sterile inflammation, which makes them good candidates to develop and improve diagnostic and therapeutic strategies in renal fibrosis and congenital obstructive nephropathies.
Collapse
Affiliation(s)
- Maja Wyczanska
- Department of Pediatrics, Dr. v. Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Bärbel Lange-Sperandio
- Department of Pediatrics, Dr. v. Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
7
|
RAGE/NF-κB pathway mediates hypoxia-induced insulin resistance in 3T3-L1 adipocytes. Biochem Biophys Res Commun 2020; 521:77-83. [DOI: 10.1016/j.bbrc.2019.10.076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 10/09/2019] [Indexed: 12/26/2022]
|
8
|
Blocking LFA-1 Aggravates Cardiac Inflammation in Experimental Autoimmune Myocarditis. Cells 2019; 8:cells8101267. [PMID: 31627327 PMCID: PMC6830329 DOI: 10.3390/cells8101267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/09/2019] [Accepted: 10/09/2019] [Indexed: 01/13/2023] Open
Abstract
The lymphocyte function-associated antigen 1 (LFA-1) is a member of the beta2-integrin family and plays a pivotal role for T cell activation and leukocyte trafficking under inflammatory conditions. Blocking LFA-1 has reduced or aggravated inflammation depending on the inflammation model. To investigate the effect of LFA-1 in myocarditis, mice with experimental autoimmune myocarditis (EAM) were treated with a function blocking anti-LFA-1 antibody from day 1 of disease until day 21, the peak of inflammation. Cardiac inflammation was evaluated by measuring infiltration of leukocytes into the inflamed cardiac tissue using histology and flow cytometry and was assessed by analysis of the heart weight/body weight ratio. LFA-1 antibody treatment severely enhanced leukocyte infiltration, in particular infiltration of CD11b+ monocytes, F4/80+ macrophages, CD4+ T cells, Ly6G+ neutrophils, and CD133+ progenitor cells at peak of inflammation which was accompanied by an increased heart weight/body weight ratio. Thus, blocking LFA-1 starting at the time of immunization severely aggravated acute cardiac inflammation in the EAM model.
Collapse
|
9
|
Bartling B, Zunkel K, Al-Robaiy S, Dehghani F, Simm A. Gene doubling increases glyoxalase 1 expression in RAGE knockout mice. Biochim Biophys Acta Gen Subj 2019; 1864:129438. [PMID: 31526867 DOI: 10.1016/j.bbagen.2019.129438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND The receptor for advanced glycation end-products (RAGE) is a multifunctional protein. Its function as pattern recognition receptor able to interact with various extracellular ligands is well described. Genetically modified mouse models, especially the RAGE knockout (RAGE-KO) mouse, identified the amplification of the immune response as an important function of RAGE. Pro-inflammatory ligands of RAGE are also methylglyoxal-derived advanced glycation end-products, which depend in their quantity, at least in part, on the activity of the methylglyoxal-detoxifying enzyme glyoxalase-1 (Glo1). Therefore, we studied the potential interaction of RAGE and Glo1 by use of RAGE-KO mice. METHODS Various tissues (lung, liver, kidney, heart, spleen, and brain) and blood cells from RAGE-KO and wildtype mice were analyzed for Glo1 expression and activity by biochemical assays and the Glo1 gene status by PCR techniques. RESULTS We identified an about two-fold up-regulation of Glo1 expression and activity in all tissues of RAGE-KO mice. This was result of a copy number variation of the Glo1 gene on mouse chromosome 17. In liver tissue and blood cells, the Glo1 expression and activity was additionally influenced by sex with higher values for male than female animals. As the genomic region containing Glo1 also contains the full-length sequence of another gene, namely Dnahc8, both genes were duplicated in RAGE-KO mice. CONCLUSION A genetic variance in RAGE-KO mice falsely suggests an interaction of RAGE and Glo1 function. GENERAL SIGNIFICANCE RAGE-independent up-regulation of Glo1 in RAGE-KO mice might be as another explanation for, at least some, effects attributed to RAGE before.
Collapse
Affiliation(s)
- Babett Bartling
- Department of Cardiac Surgery, Middle German Heart Center, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| | - Katja Zunkel
- Department of Cardiac Surgery, Middle German Heart Center, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Samiya Al-Robaiy
- Department of Cardiac Surgery, Middle German Heart Center, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Faramarz Dehghani
- Institute of Anatomy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Andreas Simm
- Department of Cardiac Surgery, Middle German Heart Center, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
10
|
Kranig SA, Tschada R, Braun M, Patry C, Pöschl J, Frommhold D, Hudalla H. Dystrophin deficiency promotes leukocyte recruitment in mdx mice. Pediatr Res 2019; 86:188-194. [PMID: 31091530 DOI: 10.1038/s41390-019-0427-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/11/2019] [Accepted: 05/03/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND A growing body of evidence defines inflammation as a hallmark feature of disease pathogenesis of Duchenne muscular dystrophy. To tailor potential immune modulatory interventions, a better understanding of immune dysregulation in Duchenne muscular dystrophy is needed. We now asked whether dystrophin deficiency affects the cascade of leukocyte recruitment. METHODS We performed intravital microscopy on the cremaster muscle of wild-type and dystrophin-deficient mdx mice. Recruitment was triggered by preparation alone (traumatic inflammation) or in combination with scrotal TNFα injections. Neutrophilic infiltration of the cremaster muscle was assessed on tissue sections. Integrin expression on circulating neutrophils and serum levels of pro-inflammatory cytokines were measured by flow cytometry. RESULTS Mdx mice show increased rolling and adhesion at baseline (traumatic inflammation) and a more profound response upon TNFα injection compared with wild-type animals. In both models, neutrophilic infiltration of the cremaster muscle is increased. Upregulation of the integrins LFA-1 and Mac-1 on circulating leukocytes and pro-inflammatory cytokines IL-6 and CCL2 in the serum points toward systemically altered immune regulation in mdx mice. CONCLUSION We are the first to show exaggerated activation of the leukocyte recruitment cascade in a dystrophin-deficient organism in vivo.
Collapse
Affiliation(s)
- Simon Alexander Kranig
- Department of Neonatology, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany
| | - Raphaela Tschada
- Department of Neonatology, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany
| | - Maylis Braun
- Department of Neonatology, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany
| | - Christian Patry
- Department of General Pediatrics, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany
| | - Johannes Pöschl
- Department of Neonatology, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany
| | - David Frommhold
- Klinik für Kinderheilkunde und Jugendmedizin, 87700, Memmingen, Germany
| | - Hannes Hudalla
- Department of Neonatology, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany.
| |
Collapse
|
11
|
Liu R, Li X, Wei J, Liu S, Chang Y, Zhang J, Zhang J, Zhang X, Fuhr U, Taubert M, Tian X. A Single Dose of Baicalin Has No Clinically Significant Effect on the Pharmacokinetics of Cyclosporine A in Healthy Chinese Volunteers. Front Pharmacol 2019; 10:518. [PMID: 31156436 PMCID: PMC6528491 DOI: 10.3389/fphar.2019.00518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/24/2019] [Indexed: 12/02/2022] Open
Abstract
Despite its narrow therapeutic window and large interindividual variability, cyclosporine A (CsA) is the first-line therapy following organ transplantation. Metabolized mainly by CYP3A and being a substrate of P-glycoprotein (P-gp), CsA is susceptible to drug–drug interactions. Baicalin (BG) is a drug used for adjuvant therapy of hepatitis in traditional Chinese medicine. Since its aglycone baicalein (B) inhibits CYP3A and P-gP, co-administration might affect CsA pharmacokinetics. This study investigated the effect of BG on CsA pharmacokinetics. In a two-period study, 16 healthy volunteers received a single 200 mg oral CsA dose alone (reference period) or in combination with 500 mg BG (test period). Pharmacokinetic evaluation of CsA was carried out using non-compartmental analysis (NCA) and population pharmacokinetics (popPK). Treatments were compared using the standard bioequivalence method. Based on NCA, 90% CIs of AUC and Cmax test-to-reference ratios were within bioequivalence boundaries. In the popPK analysis, a two-compartment model (clearance/F 62.8 L/h, central and peripheral volume of distribution/F 254 L and 388 L) with transit compartments for absorption appropriately described CsA concentrations. No clinically relevant effect of 500 mg BG co-administration on CsA pharmacokinetics was identified and both treatments were well tolerated.
Collapse
Affiliation(s)
- Ruijuan Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Xia Li
- Department I of Pharmacology, Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, University of Cologne, Cologne, Germany
| | - Jingyao Wei
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Shuaibing Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Yuanyuan Chang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Jiali Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Ji Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Uwe Fuhr
- Department I of Pharmacology, Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, University of Cologne, Cologne, Germany
| | - Max Taubert
- Department I of Pharmacology, Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, University of Cologne, Cologne, Germany
| | - Xin Tian
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| |
Collapse
|
12
|
Gonçalves-de-Albuquerque CF, Rohwedder I, Silva AR, Ferreira AS, Kurz ARM, Cougoule C, Klapproth S, Eggersmann T, Silva JD, de Oliveira GP, Capelozzi VL, Schlesinger GG, Costa ER, Estrela Marins RDCE, Mócsai A, Maridonneau-Parini I, Walzog B, Macedo Rocco PR, Sperandio M, de Castro-Faria-Neto HC. The Yin and Yang of Tyrosine Kinase Inhibition During Experimental Polymicrobial Sepsis. Front Immunol 2018; 9:901. [PMID: 29760707 PMCID: PMC5936983 DOI: 10.3389/fimmu.2018.00901] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/11/2018] [Indexed: 12/29/2022] Open
Abstract
Neutrophils are the first cells of our immune system to arrive at the site of inflammation. They release cytokines, e.g., chemokines, to attract further immune cells, but also actively start to phagocytose and kill pathogens. In the case of sepsis, this tightly regulated host defense mechanism can become uncontrolled and hyperactive resulting in severe organ damage. Currently, no effective therapy is available to fight sepsis; therefore, novel treatment targets that could prevent excessive inflammatory responses are warranted. Src Family tyrosine Kinases (SFK), a group of tyrosine kinases, have been shown to play a major role in regulating immune cell recruitment and host defense. Leukocytes with SFK depletion display severe spreading and migration defects along with reduced cytokine production. Thus, we investigated the effects of dasatinib, a tyrosine kinase inhibitor, with a strong inhibitory capacity on SFKs during sterile inflammation and polymicrobial sepsis in mice. We found that dasatinib-treated mice displayed diminished leukocyte adhesion and extravasation in tumor necrosis factor-α-stimulated cremaster muscle venules in vivo. In polymicrobial sepsis, sepsis severity, organ damage, and clinical outcome improved in a dose-dependent fashion pointing toward an optimal therapeutic window for dasatinib dosage during polymicrobial sepsis. Dasatinib treatment may, therefore, provide a balanced immune response by preventing an overshooting inflammatory reaction on the one side and bacterial overgrowth on the other side.
Collapse
Affiliation(s)
- Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany.,Laboratório de Imunofarmacologia, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ina Rohwedder
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | | | - Angela R M Kurz
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Sarah Klapproth
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - Tanja Eggersmann
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - Johnatas D Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gisele Pena de Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vera Luiza Capelozzi
- Laboratório de Genômica Pulmonar, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Edlaine Rijo Costa
- Laboratorio de Farmacologia, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rita de Cassia Elias Estrela Marins
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.,Laboratório de Pesquisa Clínica em DST e AIDS, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Attila Mócsai
- MTA-SE "Lendület" Inflammation Physiology Research Group, Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Barbara Walzog
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Markus Sperandio
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | | |
Collapse
|
13
|
Collins KH, Herzog W, MacDonald GZ, Reimer RA, Rios JL, Smith IC, Zernicke RF, Hart DA. Obesity, Metabolic Syndrome, and Musculoskeletal Disease: Common Inflammatory Pathways Suggest a Central Role for Loss of Muscle Integrity. Front Physiol 2018; 9:112. [PMID: 29527173 PMCID: PMC5829464 DOI: 10.3389/fphys.2018.00112] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/05/2018] [Indexed: 01/14/2023] Open
Abstract
Inflammation can arise in response to a variety of stimuli, including infectious agents, tissue injury, autoimmune diseases, and obesity. Some of these responses are acute and resolve, while others become chronic and exert a sustained impact on the host, systemically, or locally. Obesity is now recognized as a chronic low-grade, systemic inflammatory state that predisposes to other chronic conditions including metabolic syndrome (MetS). Although obesity has received considerable attention regarding its pathophysiological link to chronic cardiovascular conditions and type 2 diabetes, the musculoskeletal (MSK) complications (i.e., muscle, bone, tendon, and joints) that result from obesity-associated metabolic disturbances are less frequently interrogated. As musculoskeletal diseases can lead to the worsening of MetS, this underscores the imminent need to understand the cause and effect relations between the two, and the convergence between inflammatory pathways that contribute to MSK damage. Muscle mass is a key predictor of longevity in older adults, and obesity-induced sarcopenia is a significant risk factor for adverse health outcomes. Muscle is highly plastic, undergoes regular remodeling, and is responsible for the majority of total body glucose utilization, which when impaired leads to insulin resistance. Furthermore, impaired muscle integrity, defined as persistent muscle loss, intramuscular lipid accumulation, or connective tissue deposition, is a hallmark of metabolic dysfunction. In fact, many common inflammatory pathways have been implicated in the pathogenesis of the interrelated tissues of the musculoskeletal system (e.g., tendinopathy, osteoporosis, and osteoarthritis). Despite these similarities, these diseases are rarely evaluated in a comprehensive manner. The aim of this review is to summarize the common pathways that lead to musculoskeletal damage and disease that result from and contribute to MetS. We propose the overarching hypothesis that there is a central role for muscle damage with chronic exposure to an obesity-inducing diet. The inflammatory consequence of diet and muscle dysregulation can result in dysregulated tissue repair and an imbalance toward negative adaptation, resulting in regulatory failure and other musculoskeletal tissue damage. The commonalities support the conclusion that musculoskeletal pathology with MetS should be evaluated in a comprehensive and integrated manner to understand risk for other MSK-related conditions. Implications for conservative management strategies to regulate MetS are discussed, as are future research opportunities.
Collapse
Affiliation(s)
- Kelsey H. Collins
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Walter Herzog
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Graham Z. MacDonald
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Raylene A. Reimer
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Jaqueline L. Rios
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- CAPES Foundation, Brasilia, Brazil
| | - Ian C. Smith
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Ronald F. Zernicke
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Departments of Orthopaedic Surgery and Biomedical Engineering, School of Kinesiology, University of Michigan, Ann Arbor, MI, United States
- Department of Surgery, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - David A. Hart
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Family Practice, The Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC, Canada
- Alberta Health Services Bone and Joint Health Strategic Clinical Network, Calgary, AB, Canada
| |
Collapse
|
14
|
Oczypok EA, Perkins TN, Oury TD. All the "RAGE" in lung disease: The receptor for advanced glycation endproducts (RAGE) is a major mediator of pulmonary inflammatory responses. Paediatr Respir Rev 2017; 23:40-49. [PMID: 28416135 PMCID: PMC5509466 DOI: 10.1016/j.prrv.2017.03.012] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 02/07/2023]
Abstract
The receptor for advanced glycation endproducts (RAGE) is a pro-inflammatory pattern recognition receptor (PRR) that has been implicated in the pathogenesis of numerous inflammatory diseases. It was discovered in 1992 on endothelial cells and was named for its ability to bind advanced glycation endproducts and promote vascular inflammation in the vessels of patients with diabetes. Further studies revealed that RAGE is most highly expressed in lung tissue and spurred numerous explorations into RAGE's role in the lung. These studies have found that RAGE is an important mediator in allergic airway inflammation (AAI) and asthma, pulmonary fibrosis, lung cancer, chronic obstructive pulmonary disease (COPD), acute lung injury, pneumonia, cystic fibrosis, and bronchopulmonary dysplasia. RAGE has not yet been targeted in the lungs of paediatric or adult clinical populations, but the development of new ways to inhibit RAGE is setting the stage for the emergence of novel therapeutic agents for patients suffering from these pulmonary conditions.
Collapse
Affiliation(s)
| | | | - Tim D. Oury
- Corresponding author. Tel.: +1 412 648 9659; Fax: +1 412 648 9527
| |
Collapse
|
15
|
Oczypok EA, Milutinovic PS, Alcorn JF, Khare A, Crum LT, Manni ML, Epperly MW, Pawluk AM, Ray A, Oury TD. Pulmonary receptor for advanced glycation end-products promotes asthma pathogenesis through IL-33 and accumulation of group 2 innate lymphoid cells. J Allergy Clin Immunol 2015; 136:747-756.e4. [PMID: 25930197 PMCID: PMC4562894 DOI: 10.1016/j.jaci.2015.03.011] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 03/04/2015] [Accepted: 03/06/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Single nucleotide polymorphisms in the human gene for the receptor for advanced glycation end-products (RAGE) are associated with an increased incidence of asthma. RAGE is highly expressed in the lung and has been reported to play a vital role in the pathogenesis of murine models of asthma/allergic airway inflammation (AAI) by promoting expression of the type 2 cytokines IL-5 and IL-13. IL-5 and IL-13 are prominently secreted by group 2 innate lymphoid cells (ILC2s), which are stimulated by the proallergic cytokine IL-33. OBJECTIVE We sought to test the hypothesis that pulmonary RAGE is necessary for allergen-induced ILC2 accumulation in the lung. METHODS AAI was induced in wild-type and RAGE knockout mice by using IL-33, house dust mite extract, or Alternaria alternata extract. RAGE's lung-specific role in type 2 responses was explored with bone marrow chimeras and induction of gastrointestinal type 2 immune responses. RESULTS RAGE was found to drive AAI by promoting IL-33 expression in response to allergen and by coordinating the inflammatory response downstream of IL-33. Absence of RAGE impedes pulmonary accumulation of ILC2s in models of AAI. Bone marrow chimera studies suggest that pulmonary parenchymal, but not hematopoietic, RAGE has a central role in promoting AAI. In contrast to the lung, the absence of RAGE does not affect IL-33-induced ILC2 influx in the spleen, type 2 cytokine production in the peritoneum, or mucus hypersecretion in the gastrointestinal tract. CONCLUSIONS For the first time, this study demonstrates that a parenchymal factor, RAGE, mediates lung-specific accumulation of ILC2s.
Collapse
Affiliation(s)
- Elizabeth A Oczypok
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Pavle S Milutinovic
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - John F Alcorn
- Department of Pediatrics, Children's Hospital of Pittsburgh, Pittsburgh, Pa
| | - Anupriya Khare
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Lauren T Crum
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Michelle L Manni
- Department of Pediatrics, Children's Hospital of Pittsburgh, Pittsburgh, Pa
| | - Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pa
| | - Adriane M Pawluk
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Anuradha Ray
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pa.
| |
Collapse
|
16
|
García-Salido A, Oñoro G, Melen GJ, Gómez-Piña V, Serrano-González A, Ramírez-Orellana M, Casado-Flores J. Serum sRAGE as a potential biomarker for pediatric bronchiolitis: a pilot study. Lung 2015; 193:19-23. [PMID: 25355250 DOI: 10.1007/s00408-014-9663-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 10/23/2014] [Indexed: 01/14/2023]
Abstract
PURPOSE Traditional inflammatory biomarkers are insufficient for the evaluation of bronchiolitis severity. Recent investigations have shown that the receptor for advanced glycation end product (RAGE) and its soluble isoforms (sRAGE) play a critical role in the pathogenesis of lung injury. Main objective was to assess the serum levels of sRAGE of children with severe bronchiolitis admitted to the pediatric intensive care unit (PICU). Secondary objective was to study sRAGE correlation with the evolution and traditional biomarkers. METHODS Prospective, observational and descriptive study, 43 healthy controls and 37 patients (December 2011-February 2012) were enrolled. sRAGE levels were assessed and compared. In patients, the relation between sRAGE levels and clinical evolution, respiratory assistance, white blood cell count, absolute neutrophils count, serum C-reactive protein, and serum procalcitonin was analyzed. RESULTS A statistical difference was found in the mean value of sRAGE at PICU admission between patients and controls (1,215.7 ± 535 vs 849 ± 579 pg/ml). Also a significant inverse correlation was found between sRAGE and the Wood-Downes Score at admission (p = 0.02). CONCLUSIONS Serum sRAGE could be elevated in children with bronchiolitis. Larger clinical studies are necessary to elucidate its role as a bronchiolitis inflammatory and/or lung injury biomarker.
Collapse
Affiliation(s)
- Alberto García-Salido
- Pediatric Critical Care Unit, Hospital Infantil Universitario Niño Jesús, Avenida Menéndez Pelayo 65, Madrid, Spain,
| | | | | | | | | | | | | |
Collapse
|
17
|
Buschmann K, Tschada R, Metzger MS, Braach N, Kuss N, Hudalla H, Poeschl J, Frommhold D. RAGE controls leukocyte adhesion in preterm and term infants. BMC Immunol 2014; 15:53. [PMID: 25428166 PMCID: PMC4256735 DOI: 10.1186/s12865-014-0053-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 11/03/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Insufficient leukocyte recruitment may be one reason for the high incidence of life-threatening infections in preterm infants. Since the receptor of advanced glycation end products (RAGE) is a known leukocyte adhesion molecule and highly expressed during early development, we asked whether RAGE plays a role for leukocyte recruitment in preterm and term infants. METHODS Leukocyte adhesion was analyzed in dynamic flow chamber experiments using isolated leukocytes of cord blood from extremely premature (<30 weeks of gestation), moderately premature (30-35 weeks of gestation) and mature neonates (>35 weeks of gestation) and compared to the results of adults. For fluorescent microscopy leukocytes were labeled with rhodamine 6G. In the respective age groups we also measured the plasma concentration of soluble RAGE (sRAGE) by ELISA and Mac-1 and LFA-1 expression on neutrophils by flow cytometry. RESULTS The adhesive functions of fetal leukocytes significantly increase with gestational age. In all age groups, leukocyte adhesion was crucially dependent on RAGE. In particular, RAGE was equally effective to mediate leukocyte adhesion when compared to ICAM-1. The plasma levels of sRAGE were high in extremely premature infants and decreased with increasing gestational age. In contrast, expression of β2-Integrins Mac-1 and LFA-1 which are known ligands for RAGE and ICAM-1 did not change during fetal development. CONCLUSION We conclude that RAGE is a crucial leukocyte adhesion molecule in both preterm and term infants.
Collapse
Affiliation(s)
- Kirsten Buschmann
- Department of Neonatology, University Hospital, 69120, Heidelberg, Germany.
| | - Raphaela Tschada
- Department of Neonatology, University Hospital, 69120, Heidelberg, Germany.
| | | | - Natascha Braach
- Department of Neonatology, University Hospital, 69120, Heidelberg, Germany.
| | - Navina Kuss
- Department of Neonatology, University Hospital, 69120, Heidelberg, Germany.
| | - Hannes Hudalla
- Department of Neonatology, University Hospital, 69120, Heidelberg, Germany.
| | - Johannes Poeschl
- Department of Neonatology, University Hospital, 69120, Heidelberg, Germany.
| | - David Frommhold
- Department of Neonatology, University Hospital, 69120, Heidelberg, Germany.
| |
Collapse
|
18
|
Anti-inflammatory functions of protein C require RAGE and ICAM-1 in a stimulus-dependent manner. Mediators Inflamm 2014; 2014:743678. [PMID: 24876676 PMCID: PMC4024424 DOI: 10.1155/2014/743678] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/18/2014] [Accepted: 04/02/2014] [Indexed: 12/30/2022] Open
Abstract
By binding β2-integrins both ICAM-1 and the receptor for advanced glycation end products (RAGE) mediate leukocyte recruitment in a stimulus-dependent manner. Using different inflammatory mouse models we investigated how RAGE and ICAM-1 are involved in anti-inflammatory functions of protein C (PC; Ceprotin, 100 U/kg). We found that, depending on the stimulus, RAGE and ICAM-1 are cooperatively involved in PC-induced inhibition of leukocyte recruitment in cremaster models of inflammation. During short-term proinflammatory stimulation (trauma, fMLP, and CXCL1), ICAM-1 is more important for mediation of anti-inflammatory effects of PC, whereas RAGE plays a major role after longer proinflammatory stimulation (TNFα). In contrast to WT and Icam-1−/− mice, PC had no effect on bronchoalveolar neutrophil emigration in RAGE−/− mice during LPS-induced acute lung injury, suggesting that RAGE critically mediates PC effects during acute lung inflammation. In parallel, PC treatment effectively blocked leukocyte recruitment and improved survival of WT mice and Icam-1-deficient mice in LPS-induced endotoxemia, but failed to do so in RAGE-deficient mice. Exploring underlying mechanisms, we found that PC is capable of downregulating intracellular RAGE and extracellular ICAM-1 in endothelial cells. Taken together, our data show that RAGE and ICAM-1 are required for the anti-inflammatory functions of PC.
Collapse
|
19
|
Milutinovic PS, Englert JM, Crum LT, Mason NS, Ramsgaard L, Enghild JJ, Sparvero LJ, Lotze MT, Oury TD. Clearance kinetics and matrix binding partners of the receptor for advanced glycation end products. PLoS One 2014; 9:e88259. [PMID: 24642901 PMCID: PMC3958346 DOI: 10.1371/journal.pone.0088259] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 01/07/2014] [Indexed: 01/11/2023] Open
Abstract
Elucidating the sites and mechanisms of sRAGE action in the healthy state is vital to better understand the biological importance of the receptor for advanced glycation end products (RAGE). Previous studies in animal models of disease have demonstrated that exogenous sRAGE has an anti-inflammatory effect, which has been reasoned to arise from sequestration of pro-inflammatory ligands away from membrane-bound RAGE isoforms. We show here that sRAGE exhibits in vitro binding with high affinity and reversibly to extracellular matrix components collagen I, collagen IV, and laminin. Soluble RAGE administered intratracheally, intravenously, or intraperitoneally, does not distribute in a specific fashion to any healthy mouse tissue, suggesting against the existence of accessible sRAGE sinks and receptors in the healthy mouse. Intratracheal administration is the only effective means of delivering exogenous sRAGE to the lung, the organ in which RAGE is most highly expressed; clearance of sRAGE from lung does not differ appreciably from that of albumin.
Collapse
Affiliation(s)
- Pavle S. Milutinovic
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Judson M. Englert
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Lauren T. Crum
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Neale S. Mason
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Lasse Ramsgaard
- Center for Insoluble Protein Structures, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J. Enghild
- Center for Insoluble Protein Structures, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Louis J. Sparvero
- Department of Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Michael T. Lotze
- Department of Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Tim D. Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
20
|
Fuentes E, Rojas A, Palomo I. Role of multiligand/RAGE axis in platelet activation. Thromb Res 2014; 133:308-314. [PMID: 24296115 DOI: 10.1016/j.thromres.2013.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/08/2013] [Accepted: 11/12/2013] [Indexed: 02/07/2023]
Abstract
In the context of plaque progression, platelet hyperactivity associated with hyperlipidemia contributes to the development of a pro-thrombotic state. In this context, it has been demonstrated that advanced glycation end products (AGEs) significantly increases platelet activation and receptor for AGEs (RAGE) expression at the platelet surface membrane. In addition to AGEs, other ligands (S100, HMGB1 and amyloid β, among others) of RAGE have raised particular attention in platelet activation. Therefore, in this article we describe platelet hyperactivity by AGEs via RAGE-independent and RAGE-dependent pathways.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Talca, Chile
| | - Armando Rojas
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca, Chile.
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Talca, Chile.
| |
Collapse
|
21
|
Chen M, Glenn JV, Dasari S, McVicar C, Ward M, Colhoun L, Quinn M, Bierhaus A, Xu H, Stitt AW. RAGE regulates immune cell infiltration and angiogenesis in choroidal neovascularization. PLoS One 2014; 9:e89548. [PMID: 24586862 PMCID: PMC3935881 DOI: 10.1371/journal.pone.0089548] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 01/23/2014] [Indexed: 12/17/2022] Open
Abstract
Purpose RAGE regulates pro-inflammatory responses in diverse cells and tissues. This study has investigated if RAGE plays a role in immune cell mobilization and choroidal neovascular pathology that is associated with the neovascular form of age-related macular degeneration (nvAMD). Methods RAGE null (RAGE−/−) mice and age-matched wild type (WT) control mice underwent laser photocoagulation to generate choroidal neovascularization (CNV) lesions which were then analyzed for morphology, S100B immunoreactivity and inflammatory cell infiltration. The chemotactic ability of bone marrow derived macrophages (BMDMs) towards S100B was investigated. Results RAGE expression was significantly increased in the retina during CNV of WT mice (p<0.001). RAGE−/− mice exhibited significantly reduced CNV lesion size when compared to WT controls (p<0.05). S100B mRNA was upregulated in the lasered WT retina but not RAGE−/− retina and S100B immunoreactivity was present within CNV lesions although levels were less when RAGE−/− mice were compared to WT controls. Activated microglia in lesions were considerably less abundant in RAGE−/− mice when compared to WT counterparts (p<0.001). A dose dependent chemotactic migration was observed in BMDMs from WT mice (p<0.05–0.01) but this was not apparent in cells isolated from RAGE−/− mice. Conclusions RAGE-S100B interactions appear to play an important role in CNV lesion formation by regulating pro-inflammatory and angiogenic responses. This study highlights the role of RAGE in inflammation-mediated outer retinal pathology.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cells, Cultured
- Chemotaxis
- Choroidal Neovascularization/immunology
- Choroidal Neovascularization/metabolism
- Choroidal Neovascularization/pathology
- Disease Models, Animal
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/physiology
- Retina/immunology
- Retina/metabolism
- Retina/pathology
- Reverse Transcriptase Polymerase Chain Reaction
- S100 Calcium Binding Protein beta Subunit/genetics
- S100 Calcium Binding Protein beta Subunit/metabolism
Collapse
Affiliation(s)
- Mei Chen
- Centre for Experimental Medicine, Queen’s University of Belfast, Belfast, United Kingdom
| | - Josephine V. Glenn
- Centre for Experimental Medicine, Queen’s University of Belfast, Belfast, United Kingdom
| | - Shilpa Dasari
- Centre for Experimental Medicine, Queen’s University of Belfast, Belfast, United Kingdom
| | - Carmel McVicar
- Centre for Experimental Medicine, Queen’s University of Belfast, Belfast, United Kingdom
| | - Michael Ward
- Centre for Experimental Medicine, Queen’s University of Belfast, Belfast, United Kingdom
| | - Liza Colhoun
- Centre for Experimental Medicine, Queen’s University of Belfast, Belfast, United Kingdom
| | - Michael Quinn
- Centre for Experimental Medicine, Queen’s University of Belfast, Belfast, United Kingdom
| | - Angelika Bierhaus
- Department of Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Heping Xu
- Centre for Experimental Medicine, Queen’s University of Belfast, Belfast, United Kingdom
| | - Alan W. Stitt
- Centre for Experimental Medicine, Queen’s University of Belfast, Belfast, United Kingdom
- * E-mail:
| |
Collapse
|
22
|
Braach N, Frommhold D, Buschmann K, Pflaum J, Koch L, Hudalla H, Staudacher K, Wang H, Isermann B, Nawroth P, Poeschl J. RAGE controls activation and anti-inflammatory signalling of protein C. PLoS One 2014; 9:e89422. [PMID: 24586767 PMCID: PMC3933550 DOI: 10.1371/journal.pone.0089422] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/21/2014] [Indexed: 12/30/2022] Open
Abstract
AIMS The receptor for advanced glycation endproducts, RAGE, is a multiligand receptor and NF-κB activator leading to perpetuation of inflammation. We investigated whether and how RAGE is involved in mediation of anti-inflammatory properties of protein C. METHODS AND RESULTS We analyzed the effect of protein C on leukocyte adhesion and transmigration in WT- and RAGE-deficient mice using intravital microscopy of cremaster muscle venules during trauma- and TNFα-induced inflammation. Both, protein C (PC, Ceprotin, 100 U/kg) and activated protein C (aPC, 24 µg/kg/h) treatment significantly inhibited leukocyte adhesion in WT mice in these inflammation models. The impaired leukocyte adhesion after trauma-induced inflammation in RAGE knockout mice could not be further reduced by PC and aPC. After TNFα-stimulation, however, aPC but not PC treatment effectively blocked leukocyte adhesion in these mice. Consequently, we asked whether RAGE is involved in PC activation. Since RAGE-deficient mice and endothelial cells showed insufficient PC activation, and since thrombomodulin (TM) and endothelial protein C receptor (EPCR) are reduced on the mRNA and protein level in RAGE deficient endothelial cells, an involvement of RAGE in TM-EPCR-dependent PC activation is likely. Moreover, TNFα-induced activation of MAPK and upregulation of ICAM-1 and VCAM-1 are reduced both in response to aPC treatment and in the absence of RAGE. Thus, there seems to be interplay of the RAGE and the PC pathway in inflammation. CONCLUSION RAGE controls anti-inflammatory properties and activation of PC, which might involve EPCR and TM.
Collapse
Affiliation(s)
- Natascha Braach
- Department of Neonatology, University Children’s Hospital, Heidelberg, Germany
| | - David Frommhold
- Department of Neonatology, University Children’s Hospital, Heidelberg, Germany
| | - Kirsten Buschmann
- Department of Neonatology, University Children’s Hospital, Heidelberg, Germany
| | - Johanna Pflaum
- Department of Neonatology, University Children’s Hospital, Heidelberg, Germany
| | - Lutz Koch
- Department of Neonatology, University Children’s Hospital, Heidelberg, Germany
| | - Hannes Hudalla
- Department of Neonatology, University Children’s Hospital, Heidelberg, Germany
| | - Kathrin Staudacher
- Department of Neonatology, University Children’s Hospital, Heidelberg, Germany
| | - Hongjie Wang
- Department of Clinical Chemistry and Biochemistry, Otto-von-Guericke-University, Magdeburg, Germany
| | - Berend Isermann
- Department of Clinical Chemistry and Biochemistry, Otto-von-Guericke-University, Magdeburg, Germany
| | - Peter Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital, Heidelberg, Germany
| | - Johannes Poeschl
- Department of Neonatology, University Children’s Hospital, Heidelberg, Germany
| |
Collapse
|
23
|
Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal 2013; 25:2185-97. [DOI: 10.1016/j.cellsig.2013.06.013] [Citation(s) in RCA: 347] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 06/15/2013] [Accepted: 06/25/2013] [Indexed: 01/03/2023]
|
24
|
Hematopoietic progenitor kinase 1 (HPK1) is required for LFA-1–mediated neutrophil recruitment during the acute inflammatory response. Blood 2013; 121:4184-94. [DOI: 10.1182/blood-2012-08-451385] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Key Points
Hematopoietic progenitor kinase 1 (HPK1) regulates LFA-1 affinity and thereby controls adhesion and postadhesion functions of neutrophils. Hematopoietic progenitor kinase 1 (HPK1) is critically involved in neutrophil trafficking during acute inflammation.
Collapse
|
25
|
RAGE-mediated interstitial fibrosis in neonatal obstructive nephropathy is independent of NF-κB activation. Kidney Int 2013; 84:911-9. [PMID: 23677242 DOI: 10.1038/ki.2013.171] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 02/10/2013] [Accepted: 02/14/2013] [Indexed: 11/08/2022]
Abstract
Urinary tract obstruction during nephron development causes tubular apoptosis, tubular atrophy, and interstitial fibrosis. Leukocyte recruitment is critical in the development of obstructive nephropathy leading to interstitial inflammation and renal fibrosis. RAGE, the receptor of advanced glycation end products, is implicated in chronic inflammation and has been recently identified as a novel receptor for the β2-integrin Mac-1, cooperating with ICAM-1 and thereby directly mediating leukocyte recruitment in vivo. Here, we studied the role of RAGE and ICAM-1 in a model of unilateral ureteral obstruction in neonatal mice. Interestingly, the number of infiltrating leukocytes was independent of RAGE and ICAM-1 in the ureteral obstructed neonatal kidney. By contrast, galectin-3, a marker for profibrogenic M2 macrophages, was strongly reduced in ureteral obstructed RAGE and RAGE-Icam1 knockout mice. Snail expression and loss of E-cadherin but not NF-κB activation were attenuated in both knockout models. Epithelial cell cycle arrest at G2/M, which mediates kidney fibrosis, and transforming growth factor-β expression were reduced in ureteral obstructed RAGE knockout mice. Thus, RAGE and ICAM-1 promote renal fibrosis in the developing kidney upon ureteral obstruction. Combined RAGE- and ICAM-1-blocking strategies may prove beneficial in neonatal obstructive nephropathy.
Collapse
|
26
|
Keratinocyte-specific deletion of the receptor RAGE modulates the kinetics of skin inflammation in vivo. J Invest Dermatol 2013; 133:2400-2406. [PMID: 23594597 DOI: 10.1038/jid.2013.185] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 02/26/2013] [Accepted: 03/15/2013] [Indexed: 02/07/2023]
Abstract
The receptor for advanced glycation end products (RAGE) is a pattern recognition receptor causally related to the pathogenesis of acute and chronic inflammation. In a mouse model of inflammation-driven skin carcinogenesis, RAGE deletion conferred protection from the development of skin tumors due to a severely impaired cutaneous inflammation. Although the impact of RAGE expression in immune cells was shown to be essential for the maintenance of a cutaneous inflammatory reaction, the role of RAGE in keratinocytes remained unsolved. Using mice harboring a keratinocyte-specific deletion of RAGE, we analyzed its role in the regulation of an acute inflammatory response that was induced by topical treatment of the back skin with the phorbol ester 12-O-tetradecanoyl-phorbol-13-acetate (TPA). We show that RAGE expression in cutaneous keratinocytes modulates the strength and kinetics of acute inflammation and supports the maintenance of epidermal keratinocyte activation. To address the underlying molecular mechanism, we isolated interfollicular epidermis by laser microdissection for gene expression analysis, and identified RAGE as a regulator in the temporal control of TPA-induced epidermal tumor necrosis factor alpha transcript levels. In summary, our data demonstrate that RAGE expression in keratinocytes is critically involved in the perpetuation of acute inflammation and support the central role of RAGE in paracrine communication between keratinocytes and stromal immune cells.
Collapse
|
27
|
Abstract
RAGE is a key molecule in the onset and sustainment of the inflammatory response. New studies indicate that RAGE might represent a new link between the innate and adaptive immune system. RAGE belongs to the superfamily of Ig cell-surface receptors and is expressed on all types of leukocytes promoting activation, migration, or maturation of the different cells. RAGE expression is prominent on the activated endothelium, where it mediates leukocyte adhesion and transmigration. Moreover, proinflammatory molecules released from the inflamed or injured vascular system induce migration and proliferation of SMCs. RAGE binds a large number of different ligands and is therefore considered as a PRR, recognizing a structural motif rather than a specific ligand. In this review, we summarize the current knowledge about the signaling pathways activated in the different cell types and discuss a potential activation mechanism of RAGE, as well as putative options for therapeutic intervention.
Collapse
Affiliation(s)
- Katrin Kierdorf
- Department of Neuropathology, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
28
|
Abstract
In adult mammals, leukocyte recruitment follows a well-defined cascade of adhesion events enabling leukocytes to leave the circulatory system and transmigrate into tissue. Currently, it is unclear whether leukocyte recruitment proceeds in a similar fashion during fetal development. Considering the fact that the incidence of neonatal sepsis increases dramatically with decreasing gestational age in humans, we hypothesized that leukocyte recruitment may be acquired only late during fetal ontogeny. To test this, we developed a fetal intravital microscopy model in pregnant mice and, using LysEGFP (neutrophil reporter) mice, investigated leukocyte recruitment during fetal development. We show that fetal blood neutrophils acquire the ability to roll and adhere on inflamed yolk sac vessels during late fetal development, whereas at earlier embryonic stages (before day E15), rolling and adhesion were essentially absent. Accordingly, flow chamber experiments showed that fetal EGFP(+) blood cells underwent efficient adhesion only when they were harvested on or after E15. Fluorescence-activated cell sorter analysis on EGFP(+) fetal blood cells revealed that surface expression of CXCR2 and less pronounced P-selectin glycoprotein ligand-1 (PSGL-1) begin to increase only late in fetal life. Taken together, our findings demonstrate that inflammation-induced leukocyte recruitment is ontogenetically regulated and enables efficient neutrophil trafficking only during late fetal life.
Collapse
|
29
|
Schmidt S, Moser M, Sperandio M. The molecular basis of leukocyte recruitment and its deficiencies. Mol Immunol 2012; 55:49-58. [PMID: 23253941 DOI: 10.1016/j.molimm.2012.11.006] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/05/2012] [Accepted: 11/05/2012] [Indexed: 12/19/2022]
Abstract
The innate immune system responds to inflammation, infection and injury by recruiting neutrophils and other leukocytes. These cells are able to leave the intravascular compartment in a process called leukocyte recruitment. This process involves several distinct steps: selectin-mediated rolling, firm adhesion via integrins, postarrest modifications including adhesion strengthening and leukocyte crawling and finally transmigration into tissue. Genetic defects affecting the different steps of the cascade can result in severe impairment in leukocyte recruitment. So far, three leukocyte adhesion deficiencies (LAD I-III) have been described in humans. These LADs are rare autosomal recessive inherited disorders and, although clinically distinct, exhibit several common features including recurrent bacterial infections and leukocytosis. In LAD-I, mutations within the β2-integrin gene result in a severe defect in β2 integrin-mediated firm leukocyte adhesion. Defects in the posttranslational fucosylation of selectin ligands dramatically reduce leukocyte rolling and lead to LAD-II. Finally, LAD-III, also known as LAD-I variant, is caused by impaired integrin activation due to mutations within the kindlin-3 gene. This review provides an overview on the molecular basis of leukocyte adhesion and its deficiencies.
Collapse
Affiliation(s)
- Sarah Schmidt
- Walter Brendel Center for Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | | | | |
Collapse
|
30
|
CXCL1-triggered interaction of LFA1 and ICAM1 control glucose-induced leukocyte recruitment during inflammation in vivo. Mediators Inflamm 2012; 2012:739176. [PMID: 23093821 PMCID: PMC3474340 DOI: 10.1155/2012/739176] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 08/16/2012] [Accepted: 08/23/2012] [Indexed: 12/26/2022] Open
Abstract
It is well acknowledged that proinflammatory stimulation during acute hyperglycemia is able to aggravate inflammatory diseases. However, the mechanisms of proinflammatory effects of glucose are controversially discussed. We investigated leukocyte recruitment after intravenous injection of glucose in different inflammatory models using intravital microscopy. Flow chamber experiments, expression analysis, functional depletion, and knockout of key adhesion molecules gave mechanistic insight in involved pathways. We demonstrated that a single injection of glucose rapidly increased blood glucose levels in a dose-dependent manner. Notably, during tumor necrosis factor (TNF) α-induced inflammation leukocyte recruitment was not further enhanced by glucose administration, whereas glucose injection profoundly augmented leukocyte adhesion and transmigration into inflamed tissue in the trauma model, indicating that proinflammatory properties of glucose are stimulus dependent. Experiments with functional or genetic inhibition of the chemokine receptor CXCR2, intercellular adhesion molecule 1 (ICAM1), and lymphocyte function antigen 1 (LFA1) suggest that keratino-derived-chemokine CXCL1-triggered interactions of ICAM1 and LFA1 are crucially involved in the trauma model of inflammation. The lacking effect of glucose on β(2) integrin expression and on leukocyte adhesion in dynamic flow chamber experiments argues against leukocyte-driven underlying mechanisms and favours an endothelial pathway since endothelial ICAM1 expression was significantly upregulated in response to glucose.
Collapse
|