51
|
Wang Z, Li B, Jiang H, Ma Y, Bao Y, Zhu X, Xia H, Jin Y. IL-8 exacerbates alcohol-induced fatty liver disease via the Akt/HIF-1α pathway in human IL-8-expressing mice. Cytokine 2020; 138:155402. [PMID: 33352397 DOI: 10.1016/j.cyto.2020.155402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 12/16/2022]
Abstract
Alcoholic fatty liver disease (AFLD) is a disease that causes liver damage due to chronic heavy drinking. AFLD is related to lipid accumulation in liver cells caused by alcohol intake. Interleukin-8 (IL-8) is an inflammatory cytokine associated with chemotaxis (deletion in mice) that has robust effects on the occurrence and development of disease by activating related signal transduction pathways to promote inflammation and cell proliferation. There is significantly increased IL-8 expression in liver disease, which may be related to the pathogenesis of AFLD. In this study, we used hydrodynamic injection to deliver the liver-specific expression vector pLIVE-hIL-8 into mice. We found that hIL-8 can exacerbate alcohol-induced fatty liver disease via the Akt/HIF-1α pathway. Exacerbated liver lipid degeneration in mice, which is characterized by excessive accumulation of triglycerides, and liver damage markers were significantly increased. Moreover, hIL-8 could increase the alcohol-induced release of ROS in fatty liver caused by alcohol and exacerbate fatty liver disease. The expression of liver lipid metabolism-related gene sterol regulatory element-binding protein-1c (SREBP-1c) was increased. Furthermore, the expression of peroxisome proliferator-activated receptor alpha (PPARα), which is related to liver fatty acid oxidation, was decreased. The findings obtained in this study of hIL-8 will help identify a potential target for the clinical treatment of AFLD.
Collapse
Affiliation(s)
- Zhihao Wang
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; Institute for Liver Diseases of Anhui Medical University, 230032 Hefei, China
| | - Biao Li
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; Institute for Liver Diseases of Anhui Medical University, 230032 Hefei, China
| | - Haiyan Jiang
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; Institute for Liver Diseases of Anhui Medical University, 230032 Hefei, China
| | - Yuchen Ma
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; Institute for Liver Diseases of Anhui Medical University, 230032 Hefei, China
| | - Yanni Bao
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; Institute for Liver Diseases of Anhui Medical University, 230032 Hefei, China
| | - Xiangyu Zhu
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, 230032 Hefei, China
| | - Hongguang Xia
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, 230032 Hefei, China
| | - Yong Jin
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 230032 Hefei, China; Institute for Liver Diseases of Anhui Medical University, 230032 Hefei, China.
| |
Collapse
|
52
|
Wang X, Gui J. Cell-mediated immunity to SARS-CoV-2. Pediatr Investig 2020; 4:281-291. [PMID: 33376956 PMCID: PMC7768298 DOI: 10.1002/ped4.12228] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viruses spread unscrupulously virtually every corner on the planet in a very quick speed leading to an unprecedented world pandemic of COVID-19 claiming a great many of people's life. Paramount importance has been given to the studies on the virus itself including genomic variation and viron structure, as well as cell entry pathway and tissue residence. Other than that, to learn the main characteristic of host immunity responding to SARS-CoV-2 infection is an eminent task for restraining virus and controlling disease progress. Beside antibody production in response to SARS-CoV-2 infection, host cellular immunity plays an indispensable role in impeding virus replication and expansion at various stages of COVID-19 disease. In this review, we summarized the recent knowledge regarding the aberrant regulation and dysfunction of multiple immune cells during SARS-CoV-2 infection. This includes the dysregulation of immune cell number, Th polarity, cytokine storm they implicated with, as well as cell function exhaustion after chronic virus stimulation. Notwithstanding that many obstacles remain to be overcome, studies on immunotherapy for COVID-19 treatment based on the known features of host immunity in response to SARS-CoV-2 infection offer us tangible benefits and hope for making this SARS-CoV-2 pandemic under control.
Collapse
Affiliation(s)
- Xiaolin Wang
- Laboratory of Tumor ImmunologyBeijing Pediatric Research InstituteBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Jingang Gui
- Laboratory of Tumor ImmunologyBeijing Pediatric Research InstituteBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| |
Collapse
|
53
|
Salminen AT, Allahyari Z, Gholizadeh S, McCloskey MC, Ajalik R, Cottle RN, Gaborski TR, McGrath JL. In vitro Studies of Transendothelial Migration for Biological and Drug Discovery. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:600616. [PMID: 35047883 PMCID: PMC8757899 DOI: 10.3389/fmedt.2020.600616] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Inflammatory diseases and cancer metastases lack concrete pharmaceuticals for their effective treatment despite great strides in advancing our understanding of disease progression. One feature of these disease pathogeneses that remains to be fully explored, both biologically and pharmaceutically, is the passage of cancer and immune cells from the blood to the underlying tissue in the process of extravasation. Regardless of migratory cell type, all steps in extravasation involve molecular interactions that serve as a rich landscape of targets for pharmaceutical inhibition or promotion. Transendothelial migration (TEM), or the migration of the cell through the vascular endothelium, is a particularly promising area of interest as it constitutes the final and most involved step in the extravasation cascade. While in vivo models of cancer metastasis and inflammatory diseases have contributed to our current understanding of TEM, the knowledge surrounding this phenomenon would be significantly lacking without the use of in vitro platforms. In addition to the ease of use, low cost, and high controllability, in vitro platforms permit the use of human cell lines to represent certain features of disease pathology better, as seen in the clinic. These benefits over traditional pre-clinical models for efficacy and toxicity testing are especially important in the modern pursuit of novel drug candidates. Here, we review the cellular and molecular events involved in leukocyte and cancer cell extravasation, with a keen focus on TEM, as discovered by seminal and progressive in vitro platforms. In vitro studies of TEM, specifically, showcase the great experimental progress at the lab bench and highlight the historical success of in vitro platforms for biological discovery. This success shows the potential for applying these platforms for pharmaceutical compound screening. In addition to immune and cancer cell TEM, we discuss the promise of hepatocyte transplantation, a process in which systemically delivered hepatocytes must transmigrate across the liver sinusoidal endothelium to successfully engraft and restore liver function. Lastly, we concisely summarize the evolving field of porous membranes for the study of TEM.
Collapse
Affiliation(s)
- Alec T. Salminen
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Zahra Allahyari
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - Shayan Gholizadeh
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - Molly C. McCloskey
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Raquel Ajalik
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Renee N. Cottle
- Bioengineering, Clemson University, Clemson, SC, United States
| | - Thomas R. Gaborski
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - James L. McGrath
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| |
Collapse
|
54
|
Pollard BS, BLANCOl JC, Pollard JR. Classical Drug Digitoxin Inhibits Influenza Cytokine Storm, With Implications for Covid-19 Therapy. In Vivo 2020; 34:3723-3730. [PMID: 33144490 PMCID: PMC7811644 DOI: 10.21873/invivo.12221] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND/AIM Influenza viruses, corona viruses and related pneumotropic viruses cause sickness and death partly by inducing cytokine storm, a hyper-proinflammatory host response by immune cells and cytokines in the host airway. Based on our in vivo experience with digitoxin as an inhibitor of TNFα-driven NFĸB signaling for cytokine expression in prostate cancer in rats and in cystic fibrosis in humans, we hypothesize that this drug will also block a virally-activated cytokine storm. Materials Methods: Digitoxin was administered intraperitoneally to cotton rats, followed by intranasal infection with 107TCID50/100 g of cotton rat with influenza strain A/Wuhan/H3N2/359/95. Daily digitoxin treatment continued until harvest on day 4 of the experiment. RESULTS The cardiac glycoside digitoxin significantly and differentially suppressed levels of the cytokines TNFα, GRO/KC, MIP2, MCP1, and IFNγ, in the cotton rat lung in the presence of influenza virus. CONCLUSION Since cytokine storm is a host response, we suggest that digitoxin may have a therapeutic potential not only for influenza and but also for coronavirus infections.
Collapse
Affiliation(s)
| | | | - John R Pollard
- Department of Neurology, University of Pennsylvania, Philadelphia PA (USA) and Christiana Care Epilepsy Center, Newark, DE, U.S.A
| |
Collapse
|
55
|
Kalchiem-Dekel O, Yao X, Barochia AV, Kaler M, Figueroa DM, Karkowsky WB, Gordon EM, Gao M, Fergusson MM, Qu X, Liu P, Li Y, Seifuddin F, Pirooznia M, Levine SJ. Apolipoprotein E Signals via TLR4 to Induce CXCL5 Secretion by Asthmatic Airway Epithelial Cells. Am J Respir Cell Mol Biol 2020; 63:185-197. [PMID: 32338995 DOI: 10.1165/rcmb.2019-0209oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The primary function of APOE (apolipoprotein E) is to mediate the transport of cholesterol- and lipid-containing lipoprotein particles into cells by receptor-mediated endocytosis. APOE also has pro- and antiinflammatory effects, which are both context and concentration dependent. For example, Apoe-/- mice exhibit enhanced airway remodeling and hyperreactivity in experimental asthma, whereas increased APOE levels in lung epithelial lining fluid induce IL-1β secretion from human asthmatic alveolar macrophages. However, APOE-mediated airway epithelial cell inflammatory responses and signaling pathways have not been defined. Here, RNA sequencing of human asthmatic bronchial brushing cells stimulated with APOE identified increased expression of mRNA transcripts encoding multiple proinflammatory genes, including CXCL5 (C-X-C motif chemokine ligand 5), an epithelial-derived chemokine that promotes neutrophil activation and chemotaxis. We subsequently characterized the APOE signaling pathway that induces CXCL5 secretion by human asthmatic small airway epithelial cells (SAECs). Neutralizing antibodies directed against TLR4 (Toll-like receptor 4), but not TLR2, attenuated APOE-mediated CXCL5 secretion by human asthmatic SAECs. Inhibition of TAK1 (transforming growth factor-β-activated kinase 1), IκKβ (inhibitor of nuclear factor κ B kinase subunit β), TPL2 (tumor progression locus 2), and JNK (c-Jun N-terminal kinase), but not p38 MAPK (mitogen-activated protein kinase) or MEK1/2 (MAPK kinase 1/2), attenuated APOE-mediated CXCL5 secretion. The roles of TAK1, IκKβ, TPL2, and JNK in APOE-mediated CXCL5 secretion were verified by RNA interference. Furthermore, RNA interference showed that after APOE stimulation, both NF-κB p65 and TPL2 were downstream of TAK1 and IκKβ, whereas JNK was downstream of TPL2. In summary, elevated levels of APOE in the airway may activate a TLR4/TAK1/IκKβ/NF-κB/TPL2/JNK signaling pathway that induces CXCL5 secretion by human asthmatic SAECs. These findings identify new roles for TLR4 and TPL2 in APOE-mediated proinflammatory responses in asthma.
Collapse
Affiliation(s)
| | - Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch
| | | | - Maryann Kaler
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch
| | | | | | | | - Meixia Gao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch
| | | | - Xuan Qu
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch
| | - Poching Liu
- DNA Sequencing and Genomics Core Facility, and
| | - Yuesheng Li
- DNA Sequencing and Genomics Core Facility, and
| | - Fayaz Seifuddin
- Bioinformatics and Computational Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mehdi Pirooznia
- Bioinformatics and Computational Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
56
|
Wu F, Chen X, Zhai L, Wang H, Sun M, Song C, Wang T, Qian Z. CXCR2 antagonist attenuates neutrophil transmigration into brain in a murine model of LPS induced neuroinflammation. Biochem Biophys Res Commun 2020; 529:839-845. [PMID: 32616311 DOI: 10.1016/j.bbrc.2020.05.124] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 05/17/2020] [Indexed: 10/24/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is a devastating neurological complication of sepsis with intolerable high motility. SAE is accompanied with brain vascular injury, endothelial hyperpermeability, and neutrophil infiltration into the brain tissue, key inflammatory processes leading to further brain edema and neuronal cell apoptosis. Recent studies from us and others suggest that the chemokine receptor C-X-C Motif Chemokine Receptor 2 (CXCR2) is crucial for neutrophil recruitment during SAE. Here we use CXCR2 antagonist SB225002 to characterize the role of CXCR2 in brain infiltration of neutrophil in a murine model of SAE. Systemic administration of high-dose LPS (10 mg/kg) induced evident neutrophil infiltration into the cerebral cortex in wild-type mice. However, CXCR2 antagonist SB225002 markedly attenuated neutrophil infiltration into brain. The CXCR2 expression on neutrophils in the peripheral circulation was dramatically downregulated in response to this LPS dose, and endothelial CXCR2 was significantly upregulated, suggesting endothelial but not neutrophil CXCR2 plays a more important role in neutrophil infiltration into brain. Strikingly, although these CXCR2 antagonist SB225002 treated mice displayed reduced neutrophil infiltration, no change in neutrophil rolling and adhesion was observed. Furthermore, we confirmed that CXCR2 agonist CXCL1 induced a marked increase in actin stress fiber synthesis and paracellular gap formation in cultured cerebral endothelial cells, which is attenuated by SB225002. Thus, these results demonstrate a selective role for endothelial CXCR2 to regulate cerebral vascular permeability and neutrophil transmigration in high-dose LPS induced neuroinflammation, and also suggest a therapeutic potential of CXCR2 antagonist SB225002 in SAE.
Collapse
Affiliation(s)
- Fengjiao Wu
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Xiaofen Chen
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Liqian Zhai
- Department of Histology and Embryology, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Hongtao Wang
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Meiqun Sun
- Department of Histology and Embryology, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Chuanwang Song
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Ting Wang
- Department of Internal Medicine, University of Arizona, Phoenix, AZ, 85004, USA.
| | - Zhongqing Qian
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, China.
| |
Collapse
|
57
|
Khan MGM, Ghosh A, Variya B, Santharam MA, Ihsan AU, Ramanathan S, Ilangumaran S. Prognostic significance of SOCS1 and SOCS3 tumor suppressors and oncogenic signaling pathway genes in hepatocellular carcinoma. BMC Cancer 2020; 20:774. [PMID: 32807134 PMCID: PMC7433106 DOI: 10.1186/s12885-020-07285-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background SOCS1 and SOCS3 genes are considered tumor suppressors in hepatocellular carcinoma (HCC) due to frequent epigenetic repression. Consistent with this notion, mice lacking SOCS1 or SOCS3 show increased susceptibility to diethylnitrosamine (DEN)-induced HCC. As SOCS1 and SOCS3 are important regulators of cytokine and growth factor signaling, their loss could activate oncogenic signaling pathways. Therefore, we examined the correlation between SOCS1/SOCS3 and key oncogenic signaling pathway genes as well as their prognostic significance in HCC. Methods The Cancer Genome Atlas dataset on HCC comprising clinical and transcriptomic data was retrieved from the cBioportal platform. The correlation between the expression of SOCS1 or SOCS3 and oncogenic pathway genes was evaluated using the GraphPad PRISM software. The inversely correlated genes were assessed for their impact on patient survival using the UALCAN platform and their expression quantified in the regenerating livers and DEN-induced HCC tissues of mice lacking Socs1 or Socs3. Finally, the Cox proportional hazards model was used to evaluate the predictive potential of SOCS1 and SOCS3 when combined with the genes of select oncogenic signaling pathways. Results SOCS1 expression was comparable between HCC and adjacent normal tissues, yet higher SOCS1 expression predicted favorable prognosis. In contrast, SOCS3 expression was significantly low in HCC, yet it lacked predictive potential. The correlation between SOCS1 or SOCS3 expression and key genes of the cell cycle, receptor tyrosine kinase, growth factor and MAPK signaling pathways were mostly positive than negative. Among the negatively correlated genes, only a few showed elevated expression in HCC and predicted survival. Many PI3K pathway genes showed mutual exclusivity with SOCS1 and/or SOCS3 and displayed independent predictive ability. Among genes that negatively correlated with SOCS1 and/or SOCS3, only CDK2 and AURKA showed corresponding modulations in the regenerating livers and DEN-induced tumors of hepatocyte-specific Socs1 or Socs3 deficient mice and predicted patient survival. The Cox proportional hazards model identified the combinations of SOCS1 or SOCS3 with CXCL8 and DAB2 as highly predictive. Conclusions SOCS1 expression in HCC has an independent prognostic value whereas SOCS3 expression does not. The predictive potential of SOCS1 expression is increased when combined with other oncogenic signaling pathway genes.
Collapse
Affiliation(s)
- Md Gulam Musawwir Khan
- Immunology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Amit Ghosh
- Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Bhavesh Variya
- Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Madanraj Appiya Santharam
- Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Awais Ullah Ihsan
- Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Sheela Ramanathan
- Immunology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada.,Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada.,CRCHUS, Sherbrooke, Québec, J1H 5N4, Canada
| | - Subburaj Ilangumaran
- Immunology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada. .,Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada. .,CRCHUS, Sherbrooke, Québec, J1H 5N4, Canada.
| |
Collapse
|
58
|
Chen WC, Chen NJ, Chen HP, Yu WK, Su VYF, Chen H, Wu HH, Yang KY. Nintedanib Reduces Neutrophil Chemotaxis via Activating GRK2 in Bleomycin-Induced Pulmonary Fibrosis. Int J Mol Sci 2020; 21:ijms21134735. [PMID: 32630825 PMCID: PMC7370174 DOI: 10.3390/ijms21134735] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/28/2022] Open
Abstract
Neutrophils are involved in the alveolitis of idiopathic pulmonary fibrosis (IPF). However, their pathogenic mechanisms are still poorly understood. Nintedanib has antifibrotic and anti-inflammatory activity in IPF. This study aimed to investigate the regulatory mechanism of nintedanib on neutrophil chemotaxis in bleomycin (BLM)-induced pulmonary fibrosis. Nintedanib was administered via oral gavage to male C57BL/6 mice 24 h after a bleomycin intratracheal injection (1.5 U/kg). Lung histopathological findings, the expression of cytokines, and the regulatory signaling pathways of neutrophil chemotaxis were analyzed. The effect of nintedanib was also investigated in a mouse model with adoptive neutrophil transfer in vivo. Nintedanib significantly decreased the histopathological changes and neutrophil recruitment in BLM-induced pulmonary fibrosis. Nintedanib mediated a downregulation of chemokine (C-X-C motif) receptor 2 (CXCR2) and very late antigen 4 (VLA-4) expression, as well as an upregulation of G protein-coupled receptor kinase 2 (GRK2) activity in peripheral blood neutrophils in BLM-induced pulmonary fibrosis. Nintedanib also decreased the activation of endothelial cells by the decreased expression of vascular cell adhesion molecule 1 (VCAM-1). The effect of nintedanib on regulating neutrophil chemotaxis was also confirmed by a mouse model with adoptive neutrophil transfer in vivo. In conclusion, nintedanib reduces neutrophil chemotaxis and endothelial cell activation to regulate the severity of BLM-induced pulmonary fibrosis. These effects are associated with an enhancement of GRK2 activity and a reduction in CXCR2 and VLA-4 expression on neutrophils and a decrease in VCAM-1 expression on endothelial cells.
Collapse
Affiliation(s)
- Wei-Chih Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (W.-C.C.); (W.-K.Y.); (H.C.); (H.-H.W.)
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (H.-P.C.); (V.Y.-F.S.)
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Nien-Jung Chen
- Institute of Microbiology and Immunology, School of Life Sciences, National Yang-Ming University, Taipei 112, Taiwan;
- Cancer Progression Research Center, National Yang-Ming University, Taipei 112, Taiwan
| | - Hsin-Pai Chen
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (H.-P.C.); (V.Y.-F.S.)
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Wen-Kuang Yu
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (W.-C.C.); (W.-K.Y.); (H.C.); (H.-H.W.)
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (H.-P.C.); (V.Y.-F.S.)
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Vincent Yi-Fong Su
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (H.-P.C.); (V.Y.-F.S.)
- Department of Internal Medicine, Taipei City Hospital, Taipei 112, Taiwan
| | - Hao Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (W.-C.C.); (W.-K.Y.); (H.C.); (H.-H.W.)
| | - Huai-Hsuan Wu
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (W.-C.C.); (W.-K.Y.); (H.C.); (H.-H.W.)
| | - Kuang-Yao Yang
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (W.-C.C.); (W.-K.Y.); (H.C.); (H.-H.W.)
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (H.-P.C.); (V.Y.-F.S.)
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Cancer Progression Research Center, National Yang-Ming University, Taipei 112, Taiwan
- Correspondence: ; Tel.: +886-2-2875-7455; Fax: +886-2-2875-7610
| |
Collapse
|
59
|
Kang KW, Lee SJ, Kim JH, Lee BH, Kim SJ, Park Y, Kim BS. Etoposide-mediated interleukin-8 secretion from bone marrow stromal cells induces hematopoietic stem cell mobilization. BMC Cancer 2020; 20:619. [PMID: 32615949 PMCID: PMC7330970 DOI: 10.1186/s12885-020-07102-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/23/2020] [Indexed: 12/25/2022] Open
Abstract
Background We assessed the mechanism of hematopoietic stem cell (HSC) mobilization using etoposide with granulocyte-colony stimulating factor (G-CSF), and determined how this mechanism differs from that induced by cyclophosphamide with G-CSF or G-CSF alone. Methods We compared the clinical features of 173 non-Hodgkin’s lymphoma patients who underwent autologous peripheral blood stem cell transplantation (auto-PBSCT). Additionally, we performed in vitro experiments to assess the changes in human bone marrow stromal cells (hBMSCs), which support the HSCs in the bone marrow (BM) niche, following cyclophosphamide or etoposide exposure. We also performed animal studies under standardized conditions to ensure the following: exclude confounding factors, mimic the conditions in clinical practice, and identify the changes in the BM niche caused by etoposide-induced chemo-mobilization or other mobilization methods. Results Retrospective analysis of the clinical data revealed that the etoposide with G-CSF mobilization group showed the highest yield of CD34+ cells and the lowest change in white blood cell counts during mobilization. In in vitro experiments, etoposide triggered interleukin (IL)-8 secretion from the BMSCs and caused long-term BMSC toxicity. To investigate the manner in which the hBMSC-released IL-8 affects hHSCs in the BM niche, we cultured hHSCs with or without IL-8, and found that the number of total, CD34+, and CD34+/CD45- cells in IL-8-treated cells was significantly higher than the respective number in hHSCs cultured without IL-8 (p = 0.014, 0.020, and 0.039, respectively). Additionally, the relative expression of CXCR2 (an IL-8 receptor), and mTOR and c-MYC (components of IL-8-related signaling pathways) increased 1 h after IL-8 treatment. In animal studies, the etoposide with G-CSF mobilization group presented higher IL-8-related cytokine and MMP9 expression and lower SDF-1 expression in the BM, compared to the groups not treated with etoposide. Conclusion Collectively, the unique mechanism of etoposide with G-CSF-induced mobilization is associated with IL-8 secretion from the BMSCs, which is responsible for the enhanced proliferation and mobilization of HSCs in the bone marrow; this was not observed with mobilization using cyclophosphamide with G-CSF or G-CSF alone. However, the long-term toxicity of etoposide toward BMSCs emphasizes the need for the development of more efficient and safe chemo-mobilization strategies.
Collapse
Affiliation(s)
- Ka-Won Kang
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University School of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Seung-Jin Lee
- Institute of Stem Cell Research, Korea University, Seoul, South Korea.,Department of Biomedical and Science, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Ji Hye Kim
- Institute of Stem Cell Research, Korea University, Seoul, South Korea.,Department of Biomedical and Science, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Byung-Hyun Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University School of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Seok Jin Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yong Park
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University School of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Byung Soo Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University School of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, South Korea. .,Institute of Stem Cell Research, Korea University, Seoul, South Korea. .,Department of Biomedical and Science, Graduate School of Medicine, Korea University, Seoul, South Korea.
| |
Collapse
|
60
|
Transcriptome Based Profiling of the Immune Cell Gene Signature in Rat Experimental Colitis and Human IBD Tissue Samples. Biomolecules 2020; 10:biom10070974. [PMID: 32610492 PMCID: PMC7407160 DOI: 10.3390/biom10070974] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/09/2020] [Accepted: 06/27/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic intestinal inflammation is characteristic of Inflammatory Bowel Disease (IBD) that is associated with the exaggerated infiltration of immune cells. A complex interplay of inflammatory mediators and different cell types in the colon are responsible for the maintenance of tissue homeostasis and affect pathological conditions. Gene expression alteration of colon biopsies from IBD patients and an in vivo rat model of colitis were examined by RNA-Seq and QPCR, while we used in silico methods, such as Ingenuity Pathway Analysis (IPA) application and the Immune Gene Signature (ImSig) package of R, to interpret whole transcriptome data and estimate immune cell composition of colon tissues. Transcriptome profiling of in vivo colitis model revealed the most significant activation of signaling pathways responsible for leukocyte recruitment and diapedesis. We observed significant alteration of genes related to glycosylation or sensing of danger signals and pro- and anti-inflammatory cytokines and chemokines, as well as adhesion molecules. We observed the elevated expression of genes that implies the accumulation of monocytes, macrophages, neutrophils and B cells in the inflamed colon tissue. In contrast, the rate of T-cells slightly decreased in the inflamed regions. Interestingly, natural killer and plasma cells do not show enrichment upon colon inflammation. In general, whole transcriptome analysis of the in vivo experimental model of colitis with subsequent bioinformatics analysis provided a better understanding of the dynamic changes in the colon tissue of IBD patients.
Collapse
|
61
|
Banakh I, Cheshire P, Rahman M, Carmichael I, Jagadeesan P, Cameron NR, Cleland H, Akbarzadeh S. A Comparative Study of Engineered Dermal Templates for Skin Wound Repair in a Mouse Model. Int J Mol Sci 2020; 21:ijms21124508. [PMID: 32630398 PMCID: PMC7350005 DOI: 10.3390/ijms21124508] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
Engineered dermal templates have revolutionised the repair and reconstruction of skin defects. Their interaction with the wound microenvironment and linked molecular mediators of wound repair is still not clear. This study investigated the wound bed and acellular "off the shelf" dermal template interaction in a mouse model. Full-thickness wounds in nude mice were grafted with allogenic skin, and either collagen-based or fully synthetic dermal templates. Changes in the wound bed showed significantly higher vascularisation and fibroblast infiltration in synthetic grafts when compared to collagen-based grafts (P ≤ 0.05). Greater tissue growth was associated with higher prostaglandin-endoperoxide synthase 2 (Ptgs2) RNA and cyclooxygenase-2 (COX-2) protein levels in fully synthetic grafts. Collagen-based grafts had higher levels of collagen III and matrix metallopeptidase 2. To compare the capacity to form a double layer skin substitute, both templates were seeded with human fibroblasts and keratinocytes (so-called human skin equivalent or HSE). Mice were grafted with HSEs to test permanent wound closure with no further treatment required. We found the synthetic dermal template to have a significantly greater capacity to support human epidermal cells. In conclusion, the synthetic template showed advantages over the collagen-based template in a short-term mouse model of wound repair.
Collapse
Affiliation(s)
- Ilia Banakh
- Skin Bioengineering Laboratory, Victorian Adult Burns Service, Alfred Health, 89 Commercial Road, Melbourne VIC 3004, Australia; (I.B.); (P.C.); (M.R.); (H.C.)
- Department of Surgery, Monash University, 99 Commercial Road, Melbourne VIC 3004, Australia
| | - Perdita Cheshire
- Skin Bioengineering Laboratory, Victorian Adult Burns Service, Alfred Health, 89 Commercial Road, Melbourne VIC 3004, Australia; (I.B.); (P.C.); (M.R.); (H.C.)
- Department of Surgery, Monash University, 99 Commercial Road, Melbourne VIC 3004, Australia
| | - Mostafizur Rahman
- Skin Bioengineering Laboratory, Victorian Adult Burns Service, Alfred Health, 89 Commercial Road, Melbourne VIC 3004, Australia; (I.B.); (P.C.); (M.R.); (H.C.)
- Department of Surgery, Monash University, 99 Commercial Road, Melbourne VIC 3004, Australia
| | - Irena Carmichael
- Monash Micro Imaging, Monash University, 99 Commercial Road, Melbourne VIC 3004, Australia;
| | - Premlatha Jagadeesan
- Material Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton VIC 3800, Australia; (P.J.); (N.R.C.)
| | - Neil R. Cameron
- Material Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton VIC 3800, Australia; (P.J.); (N.R.C.)
| | - Heather Cleland
- Skin Bioengineering Laboratory, Victorian Adult Burns Service, Alfred Health, 89 Commercial Road, Melbourne VIC 3004, Australia; (I.B.); (P.C.); (M.R.); (H.C.)
- Department of Surgery, Monash University, 99 Commercial Road, Melbourne VIC 3004, Australia
| | - Shiva Akbarzadeh
- Skin Bioengineering Laboratory, Victorian Adult Burns Service, Alfred Health, 89 Commercial Road, Melbourne VIC 3004, Australia; (I.B.); (P.C.); (M.R.); (H.C.)
- Department of Surgery, Monash University, 99 Commercial Road, Melbourne VIC 3004, Australia
- Correspondence: ; Tel.: +61-3-9903-0616
| |
Collapse
|
62
|
De Cunto G, Cavarra E, Bartalesi B, Lucattelli M, Lungarella G. Innate Immunity and Cell Surface Receptors in the Pathogenesis of COPD: Insights from Mouse Smoking Models. Int J Chron Obstruct Pulmon Dis 2020; 15:1143-1154. [PMID: 32547002 PMCID: PMC7246326 DOI: 10.2147/copd.s246219] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/03/2020] [Indexed: 12/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is mainly associated with smoking habit. Inflammation is the major initiating process whereby neutrophils and monocytes are attracted into the lung microenvironment by external stimuli present in tobacco leaves and in cigarette smoke, which promote chemotaxis, adhesion, phagocytosis, release of superoxide anions and enzyme granule contents. A minority of smokers develops COPD and different molecular factors, which contribute to the onset of the disease, have been put forward. After many years of research, the pathogenesis of COPD is still an object of debate. In vivo models of cigarette smoke-induced COPD may help to unravel cellular and molecular mechanisms underlying the pathogenesis of COPD. The mouse represents the most favored animal choice with regard to the study of immune mechanisms due to its genetic and physiological similarities to humans, the availability of a large variability of inbred strains, the presence in the species of several genetic disorders analogous to those in man, and finally on the possibility to create models “made-to-measure” by genetic manipulation. The review outlines the different response of mouse strains to cigarette smoke used in COPD studies while retaining a strong focus on their relatability to human patients. These studies reveal the importance of innate immunity and cell surface receptors in the pathogenesis of pulmonary injury induced by cigarette smoking. They further advance the way in which we use wild type or genetically manipulated strains to improve our overall understanding of a multifaceted disease such as COPD. The structural and functional features, which have been found in the different strains of mice after chronic exposure to cigarette smoke, can be used in preclinical studies to develop effective new therapeutic agents for the different phenotypes in human COPD.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
63
|
Cervantes-García D, Bahena-Delgado AI, Jiménez M, Córdova-Dávalos LE, Ruiz-Esparza Palacios V, Sánchez-Alemán E, Martínez-Saldaña MC, Salinas E. Glycomacropeptide Ameliorates Indomethacin-Induced Enteropathy in Rats by Modifying Intestinal Inflammation and Oxidative Stress. Molecules 2020; 25:molecules25102351. [PMID: 32443501 PMCID: PMC7287897 DOI: 10.3390/molecules25102351] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/18/2022] Open
Abstract
Nonsteroidal anti-inflammatory drug (NSAID)-induced enteropathy is considered a serious and increasing clinical problem without available treatment. Glycomacropeptide (GMP) is a 64-amino acid peptide derived from milk κ-casein with numerous biological activities. The aim of this study was to investigate the protective effect of GMP on NSAID enteropathy in rats. Enteropathy was induced by seven days oral indomethacin administration. Rats were orally GMP treated from seven days previous and during the establishment of the enteropathy model. Changes in metabolism, hematological and biochemical blood alterations, intestinal inflammation and oxidative damage were analyzed. Integrity barrier markers, macroscopic intestinal damage and survival rate were also evaluated. GMP treatment prevented anorexia and weight loss in animals. Furthermore, prophylaxis with GMP ameliorated the decline in hemoglobin, hematocrit, albumin and total protein levels. The treatment had no therapeutic efficacy on the decrease of occludin and mucin (MUC)-2 expression in intestinal tissue. However, GMP markedly decreased neutrophil infiltration, and CXCL1, interleukin-1β and inducible nitric oxide synthase expression. Nitric oxide production and lipid hydroperoxide level in the small intestine were also diminished. These beneficial effects were mirrored by preventing ulcer development and increasing animal survival. These results suggest that GMP may protect against NSAID enteropathy through anti-inflammatory and antioxidant properties.
Collapse
Affiliation(s)
- Daniel Cervantes-García
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
- National Council of Science and Technology, Mexico City 03940, Mexico
| | - Armida I. Bahena-Delgado
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
| | - Mariela Jiménez
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
| | - Laura E. Córdova-Dávalos
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
| | - Vanessa Ruiz-Esparza Palacios
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
| | - Esperanza Sánchez-Alemán
- Department of Morphology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (E.S.-A.); (M.C.M.-S.)
- Unit of Familiar Medicine #8, Mexican Institute of Social Security, Aguascalientes 20180, Mexico
| | - María C. Martínez-Saldaña
- Department of Morphology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (E.S.-A.); (M.C.M.-S.)
| | - Eva Salinas
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
- Correspondence: ; Tel.: +52-449-910-8424
| |
Collapse
|
64
|
A novel approach to treatment of thromboembolic stroke in mice: Redirecting neutrophils toward a peripherally implanted CXCL1-soaked sponge. Exp Neurol 2020; 330:113336. [PMID: 32360283 DOI: 10.1016/j.expneurol.2020.113336] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/30/2020] [Accepted: 04/28/2020] [Indexed: 11/23/2022]
Abstract
Neutrophils are considered key participants in post-ischemic stroke inflammation. They are the first white blood cells to arrive in ischemic brain and their presence in the brain tissue positively correlates with post-ischemic injury severity. CXCL1 is a neutrophil attractant chemokine and the present study evaluates whether redirecting neutrophil migration using a peripherally implanted CXCL1-soaked sponge can reduce brain inflammation and improve outcomes in a novel mouse model of thromboembolic (TE) stroke. TE stroke was induced by injection of a platelet-rich microemboli suspension into the internal carotid artery of adult C57BL/6 male mice. The model induced neuroinflammation that was associated with increases in multiple brain and serum cytokines/chemokines at the mRNA and protein levels, including very marked increases in CXCL1. In other groups of animals, an absorbable sterile hemostatic sponge, previously immersed in either saline (0.9%NaCl) or CXCL1, was implanted into subcutaneous pockets formed in the inguinal region on the left and right side following stroke surgery. Mice implanted with the sponge soaked with CXCL1 had significantly reduced neuroinflammation and infarct size after TE stroke compared to mice implanted with the sponge soaked with 0.9%NaCl. There was also reduced mortality and improved neurological deficits in the TE stroke + CXCL1 sponge group compared to the TE stroke +0.9%NaCl sponge group. In conclusion: redirecting bloodstream leukocytes toward a peripherally-implanted neutrophil chemokine CXCL1-soaked sponge improves outcomes in a novel mouse model of thromboembolic stroke. The present findings suggest a novel therapeutic strategy for patients with acute stroke.
Collapse
|
65
|
Cho KH, Choi JI, Kim JO, Jung JE, Kim DW, Kim M. Therapeutic mechanism of cord blood mononuclear cells via the IL-8-mediated angiogenic pathway in neonatal hypoxic-ischaemic brain injury. Sci Rep 2020; 10:4446. [PMID: 32157146 PMCID: PMC7064601 DOI: 10.1038/s41598-020-61441-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/25/2020] [Indexed: 01/08/2023] Open
Abstract
In a clinical trial of cerebral palsy, the level of plasma interleukin-8 (IL-8) was increased, correlated with motor improvement, after human umbilical cord blood mononuclear cell (hUCBC) infusion. This study aimed to elucidate the role of IL-8 in the therapeutic effects of hUCBCs in a mouse model of hypoxic-ischaemic brain injury (HI). In P7 HI mouse brains, hUCBC administration at day 7 after HI upregulated the gene expression of Cxcl2, the mouse IL-8 homologue and increased the expression of its receptor, CXCR2. hUCBC administration restored the sequential downstream signalling axis of p-p38/p-MAPKAPK2, NFκB, and angiogenic factors, which were downregulated by HI. An in vitro assay revealed the downregulation of the angiogenic pathway by CXCR2 knockdown and p38 inhibition. In vivo p38 inhibition prior to hUCBC administration in HI mouse brains produced identical results. Behavioural outcomes revealed a therapeutic effect (ps < 0.01) of hUCBC or IL-8 administration, which was correlated with decreases in infarct size and angiogenic findings in the striatum. In conclusion, the response of the host to hUCBC administration in mice upregulated Cxcl2, which led to the activation of the IL-8-mediated p-p38 signalling pathway. The upregulation of the downstream pathway and angiogenic growth factors via NFκB can be inferred to be the potential therapeutic mechanism of hUCBCs.
Collapse
Affiliation(s)
- Kye Hee Cho
- Department of Rehabilitation Medicine, CHA Gumi Medical Center, CHA University College of Medicine, Gumi, Gyeongsangbukdo, Republic of Korea
| | - Jee In Choi
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam, Republic of Korea
| | - Jin-Ock Kim
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, Republic of Korea
| | - Joo Eun Jung
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Dong-Wook Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - MinYoung Kim
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam, Republic of Korea. .,Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University College of Medicine, Seongnam, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
66
|
Muniroh M, Gumay AR, Indraswari DA, Bahtiar Y, Hardian H, Bakri S, Maharani N, Karlowee V, Koriyama C, Yamamoto M. Activation of MIP-2 and MCP-5 Expression in Methylmercury-Exposed Mice and Their Suppression by N-Acetyl-L-Cysteine. Neurotox Res 2020; 37:827-834. [PMID: 32040762 DOI: 10.1007/s12640-020-00174-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 01/15/2020] [Accepted: 02/02/2020] [Indexed: 01/11/2023]
Abstract
Methylmercury (MeHg) is a well-known neurotoxin of the central nervous system (CNS). Neuroinflammation is one of the main pathways of MeHg-induced CNS impairment. This study aims to investigate the expressions of IL-6, MIP-2, and MCP-5, as biomarkers in relation with MeHg-induced CNS impairment and N-acetyl-L-cysteine (NAC) treatment in mice, as well as histopathological changes of brain tissue and clinical symptom such as ataxia. Twenty male Balb/c mice, aged 8-9 weeks, were divided into 4 groups and treated with saline (control), NAC [150 mg/kg body weight (BW) day], MeHg (4 mg Hg/kg BW), or a combination of MeHg and NAC for 17 days. MeHg induced the expression of IL-6, MIP-2, and MCP-5 in the serum, with median values (those in controls) of 55.06 (9.44), 15.94 (9.30), and 458.91 (239.91) mg/dl, respectively, and a statistical significance was observed only in IL-6 expression (p < 0.05). MIP-2 and MCP-5 expressions tended to increase in the cerebrum of MeHg-treated group compared with controls; however, the difference was not statistically significant. MeHg treatment also increased IL-6 expression in the cerebellum (7.73 and 4.81 mg/dl in MeHg-treated group and controls, respectively), with a marginal significance. NAC significantly suppressed MeHg-induced IL-6 and MIP-2 expressions in the serum (p < 0.05 for both), and slightly reduced MCP-5 expression in the cerebrum. Ataxia was observed in all MeHg-treated mice after 9-day exposure as well as the decrease of intact Purkinje cells in brain tissue (p < 0.05). These findings suggest that MeHg induced neurotoxicity by elevating the expression of IL-6, MIP-2, and MCP-5 and causing ataxia symptoms, and NAC reduced MeHg-mediated effects on the CNS.
Collapse
Affiliation(s)
- Muflihatul Muniroh
- Department of Physiology, Faculty of Medicine Diponegoro University, Tembalang Semarang, 50275, Indonesia.
| | - Ainun Rahmasari Gumay
- Department of Physiology, Faculty of Medicine Diponegoro University, Tembalang Semarang, 50275, Indonesia
| | - Darmawati Ayu Indraswari
- Department of Physiology, Faculty of Medicine Diponegoro University, Tembalang Semarang, 50275, Indonesia
| | - Yuriz Bahtiar
- Department of Physiology, Faculty of Medicine Diponegoro University, Tembalang Semarang, 50275, Indonesia
| | - Hardian Hardian
- Department of Physiology, Faculty of Medicine Diponegoro University, Tembalang Semarang, 50275, Indonesia
| | - Saekhol Bakri
- Department of Public Health, Faculty of Medicine Diponegoro University, Semarang, 50275, Indonesia
| | - Nani Maharani
- Department of Pharmacology and Therapy, Faculty of Medicine Diponegoro University, Semarang, 50275, Indonesia
| | - Vega Karlowee
- Department of Anatomical Pathology, Faculty of Medicine Diponegoro University, Semarang, 50275, Indonesia
| | - Chihaya Koriyama
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Health Sciences, Kagoshima, 890-8520, Japan
| | - Megumi Yamamoto
- Department of Environment and Public Health, National Institute for Minamata Disease, Kumamoto, 867-0008, Japan
| |
Collapse
|
67
|
CD14 and ALPK1 Affect Expression of Tight Junction Components and Proinflammatory Mediators upon Bacterial Stimulation in a Colonic 3D Organoid Model. Stem Cells Int 2020; 2020:4069354. [PMID: 32076438 PMCID: PMC7016478 DOI: 10.1155/2020/4069354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/20/2019] [Accepted: 01/11/2020] [Indexed: 12/19/2022] Open
Abstract
Cd14 and Alpk1 both encode pathogen recognition receptors and are known candidate genes for affecting severity in inflammatory bowel diseases. CD14 acts as a coreceptor for bacterial lipopolysaccharide (LPS), while ALPK1 senses ADP-D-glycero-beta-D-manno-heptose, a metabolic intermediate of LPS biosynthesis. Intestinal barrier integrity can be influenced by CD14, whereas to date, the role of ALPK1 in maintaining barrier function remains unknown. We used colon-derived 3D organoids, first characterised for growth, proliferation, stem cell markers, and expression of tight junction (TJ) components using qPCR and immunohistochemistry. They showed characteristic crypt stem cells, apical shedding of dead cells, and TJ formation. Afterwards, organoids of different genotypes (WT, Il10−/−, Cd14−/−, and Alpk1−/−) were then stimulated with either LPS or Escherichia coli Nissle 1917 (EcN). Gene expression and protein levels of cytokines and TJ components were analysed. WT organoids increased expression of Tnfα and tight junction components. Cd14−/− organoids expressed significantly less Tnfα and Ocln after LPS stimulation than WT organoids but reacted similarly to WT organoids after EcN stimulation. In contrast, compared to WT, Alpk1−/− organoids showed decreased expression of different TJ and cytokine genes in response to EcN but not LPS. However, Western blotting revealed an effect of ALPK1 on TJ protein levels. These findings demonstrate that Cd14, but not Alpk1, alters the response to LPS stimulation in colonic epithelial cells, whereas Alpk1 is involved in the response upon bacterial challenge.
Collapse
|
68
|
Gamage AM, Zhu F, Ahn M, Foo RJH, Hey YY, Low DHW, Mendenhall IH, Dutertre CA, Wang LF. Immunophenotyping monocytes, macrophages and granulocytes in the Pteropodid bat Eonycteris spelaea. Sci Rep 2020; 10:309. [PMID: 31941952 PMCID: PMC6962400 DOI: 10.1038/s41598-019-57212-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 12/12/2019] [Indexed: 02/07/2023] Open
Abstract
Bats are asymptomatic reservoir hosts for several highly pathogenic viruses. Understanding this enigmatic relationship between bats and emerging zoonotic viruses requires tools and approaches which enable the comparative study of bat immune cell populations and their functions. We show that bat genomes have a conservation of immune marker genes which delineate phagocyte populations in humans, while lacking key mouse surface markers such as Ly6C and Ly6G. Cross-reactive antibodies against CD44, CD11b, CD14, MHC II, and CD206 were multiplexed to characterize circulating monocytes, granulocytes, bone-marrow derived macrophages (BMDMs) and lung alveolar macrophages (AMs) in the cave nectar bat Eonycteris spelaea. Transcriptional profiling of bat monocytes and BMDMs identified additional markers – including MARCO, CD68, CD163, CD172α, and CD88 – which can be used to further characterize bat myeloid populations. Bat cells often resembled their human counterparts when comparing immune parameters that are divergent between humans and mice, such as the expression patterns of certain immune cell markers. A genome-wide comparison of immune-related genes also revealed a much closer phylogenetic relationship between bats and humans compared to rodents. Taken together, this study provides a set of tools and a comparative framework which will be important for unravelling viral disease tolerance mechanisms in bats.
Collapse
Affiliation(s)
- Akshamal M Gamage
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Feng Zhu
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Matae Ahn
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Randy Jee Hiang Foo
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Ying Ying Hey
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Dolyce H W Low
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Ian H Mendenhall
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Charles-Antoine Dutertre
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.,Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
69
|
Westman J, Grinstein S, Marques PE. Phagocytosis of Necrotic Debris at Sites of Injury and Inflammation. Front Immunol 2020; 10:3030. [PMID: 31998312 PMCID: PMC6962235 DOI: 10.3389/fimmu.2019.03030] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022] Open
Abstract
Clearance of cellular debris is required to maintain the homeostasis of multicellular organisms. It is intrinsic to processes such as tissue growth and remodeling, regeneration and resolution of injury and inflammation. Most of the removal of effete and damaged cells is performed by macrophages and neutrophils through phagocytosis, a complex phenomenon involving ingestion and degradation of the disposable particles. The study of the clearance of cellular debris has been strongly biased toward the removal of apoptotic bodies; as a result, the mechanisms underlying the removal of necrotic cells have remained relatively unexplored. Here, we will review the incipient but growing knowledge of the phagocytosis of necrotic debris, from their recognition and engagement to their internalization and disposal. Critical insights into these events were gained recently through the development of new in vitro and in vivo models, along with advances in live-cell and intravital microscopy. This review addresses the classes of "find-me" and "eat-me" signals presented by necrotic cells and their cognate receptors in phagocytes, which in most cases differ from the extensively characterized counterparts in apoptotic cell engulfment. The roles of damage-associated molecular patterns, chemokines, lipid mediators, and complement components in recruiting and activating phagocytes are reviewed. Lastly, the physiological importance of necrotic cell removal is emphasized, highlighting the key role of impaired debris clearance in autoimmunity.
Collapse
Affiliation(s)
- Johannes Westman
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
70
|
Acker G, Zollfrank J, Jelgersma C, Nieminen-Kelhä M, Kremenetskaia I, Mueller S, Ghori A, Vajkoczy P, Brandenburg S. The CXCR2/CXCL2 signalling pathway - An alternative therapeutic approach in high-grade glioma. Eur J Cancer 2020; 126:106-115. [PMID: 31927212 DOI: 10.1016/j.ejca.2019.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 12/06/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Besides VEGF, alternative signalling via CXCR2 and its ligands CXCL2/CXCL8 is a crucial part of angiogenesis in glioblastoma. Our aim was to understand the role of CXCR2 for glioma biology and elucidate the therapeutic potential of its specific inhibition. METHODS GL261 glioma cells were implanted intracranially in syngeneic mice. The 14 or 7 days of local or systemic treatment with CXCR2-antagonist (SB225002) was initiated early on the day of tumour cell implantation or delayed after 14 days of tumour growth. Glioma volume was verified using MRI before and after treatment. Immunofluorescence staining was used to investigate tumour progression, angiogenesis and microglial behaviour. Furthermore, in vitro assays and gene expression analyses of glioma and endothelial cells were performed to validate inhibitor activity. RESULTS CXCR2-blocking led to significantly reduced glioma volumes of around 50% after early and delayed local treatments. The treated tumours were comparable with controls regarding invasiveness, proliferation and apoptotic cell activity. Furthermore, no differences in CXCR2/CXCL2 expression were observed. However, immunostaining revealed reduction in vessel density and accumulation of microglia/macrophages, whereas interaction of these myeloid cells with tumour vessels was enhanced. In vitro analyses of the CXCR2-antagonist showed its direct impact on proliferation of glioma and endothelial cells if used at higher concentrations. In addition, expression of CXCR2/CXCL2 signalling genes was increased in both cell types by SB225002, but VEGF-relevant genes were unaffected. CONCLUSION The CXCR2-antagonist inhibited glioma growth during tumour initiation and progression, whereas treatment was well-tolerated by the recipients. Thus, the CXCR2/CXCL2 signalling represents a promising therapeutic target in glioma.
Collapse
Affiliation(s)
- Güliz Acker
- Department of Neurosurgery and Experimental Neurosurgery, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany
| | - Julia Zollfrank
- Department of Neurosurgery and Experimental Neurosurgery, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Claudius Jelgersma
- Department of Neurosurgery and Experimental Neurosurgery, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Melina Nieminen-Kelhä
- Department of Neurosurgery and Experimental Neurosurgery, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Irina Kremenetskaia
- Department of Neurosurgery and Experimental Neurosurgery, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Susanne Mueller
- Department of Neurology and Experimental Neurology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Adnan Ghori
- Department of Neurosurgery and Experimental Neurosurgery, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery and Experimental Neurosurgery, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| | - Susan Brandenburg
- Department of Neurosurgery and Experimental Neurosurgery, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
71
|
The Ovarian Transcriptome of Reproductively Aged Multiparous Mice: Candidate Genes for Ovarian Cancer Protection. Biomolecules 2020; 10:biom10010113. [PMID: 31936467 PMCID: PMC7022285 DOI: 10.3390/biom10010113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 12/15/2022] Open
Abstract
In middle-aged women, the decline of ovarian follicle reserve below a critical threshold marks menopause, leading to hormonal, inflammatory, and metabolic changes linked to disease. The highest incidence and mortality of sporadic ovarian cancer (OC) occur at post-menopause, while OC risk is reduced by full-term pregnancies during former fertile life. Herein, we investigate how parity history modulates the ovarian transcriptome related to such declining follicle pool and systemic inflammation in reproductively-aged mice. Female C57BL/6 mice were housed under multiparous and virgin (nulliparous) breeding regimens from adulthood until estropause. The ovaries were then subjected to follicle count and transcriptional profiling, while a cytokine panel was determined in the sera. As expected, the follicle number was markedly decreased just by aging. Importantly, a significantly higher count of primordial and total follicles was observed in aged multiparous relative to aged virgin ovaries. Consistently, among the 65 genes of higher expression in aged multiparous ovaries, 27 showed a follicle count-like pattern, 21 had traceable evidence of roles in follicular/oocyte homeostasis, and 7 were transforming-growth factor beta (TGF-β)/bone morphogenetic protein (BMP) superfamily members. The remaining genes were enriched in cell chemotaxis and innate-immunity, and resembled the profiles of circulating CXCL1, CXCL2, CXCL5, CSF3, and CCL3, chemokines detected at higher levels in aged multiparous mice. We conclude that multiparity during reproductive life promotes the retention of follicle remnants while improving local (ovarian) and systemic immune-innate surveillance in aged female mice. These findings could underlie the mechanisms by which pregnancy promotes the long-term reduced OC risk observed at post-menopause.
Collapse
|
72
|
Particulate Matter Increases the Severity of Bleomycin-Induced Pulmonary Fibrosis through KC-Mediated Neutrophil Chemotaxis. Int J Mol Sci 2019; 21:ijms21010227. [PMID: 31905700 PMCID: PMC6981983 DOI: 10.3390/ijms21010227] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 12/26/2022] Open
Abstract
Background: Although particular matter (PM) increases incidence and severity of idiopathic pulmonary fibrosis, the underlying mechanism remains elusive. Methods: The effects of PM were evaluated in a murine model of bleomycin-induced pulmonary fibrosis. Mice were divided into four groups, receiving: (1) Saline (control), (2) bleomycin, (3) PM, or (4) bleomycin plus PM (Bleo+PM). Additional groups of Bleo+PM mice were treated with sivelestat (an inhibitor of neutrophil elastase) or reparixin (a C-X-C motif chemokine receptor 2 antagonist), or were genetically modified with keratinocyte chemoattractant (KC) deletion. Results: Pulmonary fibrosis was not observed in the control or PM groups. Bleomycin induced pulmonary fibrosis within 14 days. The Bleo+PM group showed worse pulmonary fibrosis when compared to the bleomycin group. Analyses of immune cell profile and chemokine/cytokine concentrations at day 2-bronchoalveolar lavage fluid (BALF) revealed that the Bleo+PM group had increased neutrophil number and elastase level and KC concentration compared to the bleomycin group. Neutrophil elastase activated the Smad2/Smad3/α-SMA pathway to induce collagen deposition, while sivelestat abrogated the increased severity of pulmonary fibrosis caused by PM. Chemotaxis assay revealed that BALF of the Bleo+PM group recruited neutrophil, which was dependent on KC. Further, genetic KC deletion or pharmaceutical inhibition of KC binding to CXCR2 with reparixin ameliorated the PM-induced increased severity of pulmonary fibrosis. Conclusions: These data provide evidence that the PM-induced increased severity of pulmonary fibrosis depends on KC-mediated neutrophil chemotaxis and give additional mechanic insight that will aid in the development of therapeutic strategies.
Collapse
|
73
|
Wang J, Ke Y, Shu T. Crocin has pharmacological effects against the pathological behavior of colon cancer cells by interacting with the STAT3 signaling pathway. Exp Ther Med 2019; 19:1297-1303. [PMID: 32010302 PMCID: PMC6966197 DOI: 10.3892/etm.2019.8329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 10/07/2019] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to investigate changes in proliferation, apoptosis, inflammation and chemokine release of colon cancer cells after treatment with crocin, as well as to investigate the signaling pathway that is regulated by crocin. The inhibition rates of different doses of crocin on the proliferation of HCT116 cells were measured by MTT assay. The IC50 was calculated from the inhibition rates at 48 h. Proliferation curves of HCT116 cells were plotted after treatment with 271.18 µM (high-dose group) or 135.6 µM (low-dose group) crocin. Flow cytometry and Hoechst 33342/propidium iodide double staining were used for detecting apoptosis. ELISA was used to measure the levels of macrophage inflammatory protein 2, interleukin (IL)-8, monocyte chemoattractant protein 1, tumor necrosis factor-α, IL-6 and IL-1β in the supernatant from cultured HCT116 cells following both high- and low-dose crocin treatment. Phosphorylated (P)-STAT3/STAT3 in HCT116 cells were measured by western blotting. Crocin inhibited the proliferation of HCT116 cells in a dose-dependent manner and the high-dose treatment with crocin resulted in a lower rate of proliferation. Additionally, crocin increased the apoptosis of HCT116 cells and the high-dose treatment with crocin led to a higher level of apoptosis. Notably, crocin decreased the secretion of chemokines and inflammatory factors from HCT116 cells and the high-dose treatment with crocin caused the greatest reduction in secretion of the factors. Crocin reduced the ratio of P-STAT3/STAT3, and thereby reduced the release of cytokines. The present study demonstrated that crocin may have pharmacological effects against the pathological behavior of colon cancer cells, and its mechanism of action may be related to the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Jun Wang
- Graduate School, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Yupei Ke
- Graduate School, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Tao Shu
- Department of Anorectal Surgery, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
74
|
Sasaki T, Suzuki Y, Kakisaka K, Wang T, Ishida K, Suzuki A, Abe H, Sugai T, Takikawa Y. IL-8 induces transdifferentiation of mature hepatocytes toward the cholangiocyte phenotype. FEBS Open Bio 2019; 9:2105-2116. [PMID: 31651102 PMCID: PMC6886300 DOI: 10.1002/2211-5463.12750] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/28/2019] [Accepted: 10/22/2019] [Indexed: 01/20/2023] Open
Abstract
The adult mammalian liver exhibits a remarkable regenerative capacity, with different modes of regeneration according to the type and extent of injury. Hepatocyte–cholangiocyte biphenotypic liver progenitor cell populations appear under conditions of excessive injury. It has been reported that mature hepatocytes can transdifferentiate toward a cholangiocyte phenotype and be a cellular source of progenitor cell populations. Here, we determined that among various plasma cytokines, interleukin (IL)‐8 levels were significantly elevated in acute liver failure and severe acute liver injury patients. In vitro assays revealed that administration of IL‐8 homologues increases the expression of Sry HMG box protein 9 (SOX9). In liver biopsies of acute liver injury patients, we observed the appearance of SOX9‐positive biphenotypic hepatocytes accompanied by elevation of plasma IL‐8 levels. Our results suggest that IL‐8 regulates the phenotypic conversion of mature hepatocytes toward a cholangiocyte phenotype. Interleukin (IL)‐8 treated mouse mature hepatocytes expressed Sry HMG box protein 9 (SOX9), a bile duct‐associated marker. In liver biopsies of acute liver injury patients, SOX9‐positive biphenotypic hepatocytes appeared in the liver parenchyma which is accompanied by elevation of plasma IL‐8 levels. Our results suggest that IL‐8 regulates the phenotypic conversion of mature hepatocytes toward a cholangiocyte phenotype.![]()
Collapse
Affiliation(s)
- Tokio Sasaki
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Yuji Suzuki
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Keisuke Kakisaka
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Ting Wang
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Kazuyuki Ishida
- Department of Molecular Diagnostic Pathology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Akiko Suzuki
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Hiroaki Abe
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Tamotsu Sugai
- Department of Molecular Diagnostic Pathology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Yasuhiro Takikawa
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| |
Collapse
|
75
|
Monocytes exposed to plasma from patients with Alzheimer’s disease undergo metabolic reprogramming. Neurosci Res 2019; 148:54-60. [DOI: 10.1016/j.neures.2019.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/07/2018] [Accepted: 01/09/2019] [Indexed: 12/16/2022]
|
76
|
Mendes V, Galvão I, Vieira AT. Mechanisms by Which the Gut Microbiota Influences Cytokine Production and Modulates Host Inflammatory Responses. J Interferon Cytokine Res 2019; 39:393-409. [PMID: 31013453 DOI: 10.1089/jir.2019.0011] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal tract encounters a wide variety of microorganisms, including beneficial symbionts, pathobionts, and pathogens. Recent evidence has shown that the gut microbiota, directly or indirectly through its components, such as metabolites, actively participates in the host inflammatory response by cytokine-microbiota or microbiota-cytokine modulation interactions, both in the gut and systemically. Therefore, further elucidation of host cytokine molecular pathways and microbiota components will provide a novel and promising therapeutic approach to control or prevent inflammatory disease and to maintain host homeostasis. The purpose of this review is to summarize well-established scientific findings and provide an updated overview regarding the direct and indirect mechanisms by which the gut microbiota can influence the inflammatory response by modulating the host's cytokine pathways that are mostly involved, but not exclusively so, with gut homeostasis. In addition, we will highlight recent results from our group, which suggest that the microbiota promotes cytokine release from inflammatory cells though activation of microbial metabolite sensor receptors that are more highly expressed on inflammatory and intestinal epithelial cells.
Collapse
Affiliation(s)
- Viviani Mendes
- 1 Laboratory of Microbiota and Immunomodulation, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,2 Department of General Biology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Izabela Galvão
- 3 Department of Cellular Biology ICB, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Angelica Thomaz Vieira
- 1 Laboratory of Microbiota and Immunomodulation, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,2 Department of General Biology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
77
|
Putnam NE, Fulbright LE, Curry JM, Ford CA, Petronglo JR, Hendrix AS, Cassat JE. MyD88 and IL-1R signaling drive antibacterial immunity and osteoclast-driven bone loss during Staphylococcus aureus osteomyelitis. PLoS Pathog 2019; 15:e1007744. [PMID: 30978245 PMCID: PMC6481883 DOI: 10.1371/journal.ppat.1007744] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/24/2019] [Accepted: 04/01/2019] [Indexed: 01/18/2023] Open
Abstract
Staphylococcus aureus is able to infect virtually all organ systems and is a frequently isolated etiologic agent of osteomyelitis, a common and debilitating invasive infection of bone. Treatment of osteomyelitis requires invasive surgical procedures and prolonged antibiotic therapy, yet is frequently unsuccessful due to extensive pathogen-induced bone damage that can limit antibiotic penetration and immune cell influx to the infectious focus. We previously established that S. aureus triggers profound alterations in bone remodeling in a murine model of osteomyelitis, in part through the production of osteolytic toxins. However, staphylococcal strains lacking osteolytic toxins still incite significant bone destruction, suggesting that host immune responses are also major drivers of pathologic bone remodeling during osteomyelitis. The objective of this study was to identify host immune pathways that contribute to antibacterial immunity during S. aureus osteomyelitis, and to define how these immune responses alter bone homeostasis and contribute to bone destruction. We specifically focused on the interleukin-1 receptor (IL-1R) and downstream adapter protein MyD88 given the prominent role of this signaling pathway in both antibacterial immunity and osteo-immunologic crosstalk. We discovered that while IL-1R signaling is necessary for local control of bacterial replication during osteomyelitis, it also contributes to bone loss during infection. Mechanistically, we demonstrate that S. aureus enhances osteoclastogenesis of myeloid precursors in vitro, and increases the abundance of osteoclasts residing on bone surfaces in vivo. This enhanced osteoclast abundance translates to trabecular bone loss, and is dependent on intact IL-1R signaling. Collectively, these data define IL-1R signaling as a critical component of the host response to S. aureus osteomyelitis, but also demonstrate that IL-1R-dependent immune responses trigger collateral bone damage through activation of osteoclast-mediated bone resorption.
Collapse
Affiliation(s)
- Nicole E. Putnam
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Laura E. Fulbright
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jacob M. Curry
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Caleb A. Ford
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jenna R. Petronglo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Andrew S. Hendrix
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - James E. Cassat
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Institute for Infection, Immunology and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
78
|
Pigat N, Reyes-Gomez E, Boutillon F, Palea S, Barry Delongchamps N, Koch E, Goffin V. Combined Sabal and Urtica Extracts (WS ® 1541) Exert Anti-proliferative and Anti-inflammatory Effects in a Mouse Model of Benign Prostate Hyperplasia. Front Pharmacol 2019; 10:311. [PMID: 30984003 PMCID: PMC6450068 DOI: 10.3389/fphar.2019.00311] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 03/14/2019] [Indexed: 12/19/2022] Open
Abstract
WS® 1541 is a phytopharmaceutical drug combination containing a lipophilic extract from fruits of Sabal serrulata (WS® 1473) and an aqueous ethanolic extract from roots of Urtica dioica (WS® 1031). It is approved in several countries worldwide for the treatment of lower urinary tract syndrome (LUTS) linked to benign prostate hyperplasia (BPH). Clinical studies have demonstrated the efficacy of this unique combination in the treatment of BPH-related LUTS. However, its mechanisms of action in vivo remain partly uncharacterized. The aim of this study was to take advantage of a validated mouse model of BPH to better characterize its growth-inhibitory and anti-inflammatory properties. We used the probasin–prolactin (Pb-PRL) transgenic mouse model in which prostate-specific overexpression of PRL results in several features of the human disease including tissue hypertrophy, epithelial hyperplasia, increased stromal cellularity, inflammation, and LUTS. Six-month-old heterozygous Pb-PRL male mice were randomly distributed to five groups (11–12 animals/group) orally treated for 28 consecutive days with WS® 1541 (300, 600, or 900 mg/kg/day), the 5α-reductase inhibitor finasteride used as reference (5 mg/kg/day) or vehicle (olive oil 5 ml/kg/day). Administration of WS® 1541 was well tolerated and caused a dose-dependent reduction of prostate weight (vs. vehicle) that was statistically significant at the two highest doses. This effect was accompanied by a reduction in prostate cell proliferation as assessed by lower Ki-67 expression (qPCR and immunohistochemistry). In contrast, finasteride had no or only a mild effect on these parameters. The growth-inhibitory activity of WS® 1541 was accompanied by a strong anti-inflammatory effect as evidenced by the reduced infiltration of cells expressing the leukocyte common antigen CD45. In sharp contrast, finasteride significantly increased the prostate inflammatory status according to this readout. Molecular profiling (qPCR) of 23 selected pro-inflammatory genes confirmed the strong anti-inflammatory potency of WS® 1541 compared to finasteride. Since treatment of WS® 1541 did not interfere with transgene expression and activity in the prostate of Pb-PRL mice, the effects observed in this study are entirely attributable to the intrinsic pharmacological action of the drug combination.
Collapse
Affiliation(s)
- Natascha Pigat
- PRL/GH Pathophysiology Laboratory, Institut Necker Enfants Malades, Unit 1151, Inserm, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France
| | - Edouard Reyes-Gomez
- Unité d'Histologie et d'Anatomie Pathologique, Laboratoire d'Anatomo-Cytopathologie, Biopôle Alfort, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France.,Inserm, U955 - IMRB, Ecole Nationale Vétérinaire d'Alfort, UPEC, Maisons-Alfort, France
| | - Florence Boutillon
- PRL/GH Pathophysiology Laboratory, Institut Necker Enfants Malades, Unit 1151, Inserm, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France
| | | | - Nicolas Barry Delongchamps
- PRL/GH Pathophysiology Laboratory, Institut Necker Enfants Malades, Unit 1151, Inserm, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France.,Urology Department, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Egon Koch
- Dr. Willmar Schwabe GmbH & Co. KG, Karlsruhe, Germany
| | - Vincent Goffin
- PRL/GH Pathophysiology Laboratory, Institut Necker Enfants Malades, Unit 1151, Inserm, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France
| |
Collapse
|
79
|
Milard M, Penhoat A, Durand A, Buisson C, Loizon E, Meugnier E, Bertrand K, Joffre F, Cheillan D, Garnier L, Viel S, Laugerette F, Michalski MC. Acute effects of milk polar lipids on intestinal tight junction expression: towards an impact of sphingomyelin through the regulation of IL-8 secretion? J Nutr Biochem 2019; 65:128-138. [DOI: 10.1016/j.jnutbio.2018.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/30/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
|
80
|
Carneiro-Lobo TC, Scalabrini LC, Magalhães LDS, Cardeal LB, Rodrigues FS, Dos Santos EO, Baldwin AS, Levantini E, Giordano RJ, Bassères DS. IKKβ targeting reduces KRAS-induced lung cancer angiogenesis in vitro and in vivo: A potential anti-angiogenic therapeutic target. Lung Cancer 2019; 130:169-178. [PMID: 30885340 DOI: 10.1016/j.lungcan.2019.02.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/14/2019] [Accepted: 02/25/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The ability of tumor cells to drive angiogenesis is an important cancer hallmark that positively correlates with metastatic potential and poor prognosis. Therefore, targeting angiogenesis is a rational therapeutic approach and dissecting proangiogenic pathways is important, particularly for malignancies driven by oncogenic KRAS, which are widespread and lack effective targeted therapies. Based on published studies showing that oncogenic RAS promotes angiogenesis by upregulating the proangiogenic NF-κB target genes IL-8 and VEGF, that NF-κB activation by KRAS requires the IKKβ kinase, and that targeting IKKβ reduces KRAS-induced lung tumor growth in vivo, but has limited effects on cell growth in vitro, we hypothesized that IKKβ targeting would reduce lung tumor growth by inhibiting KRAS-induced angiogenesis. MATERIALS AND METHODS To test this hypothesis, we targeted IKKβ in KRAS-mutant lung cancer cell lines either by siRNA-mediated transfection or by treatment with Compound A (CmpdA), a highly specific IKKβ inhibitor, and used in vitro and in vivo assays to evaluate angiogenesis. RESULTS AND CONCLUSIONS Both pharmacological and siRNA-mediated IKKβ targeting in lung cells reduced expression and secretion of NF-κB-regulated proangiogenic factors IL-8 and VEGF. Moreover, conditioned media from IKKβ-targeted lung cells reduced human umbilical vein endothelial cell (HUVEC) migration, invasion and tube formation in vitro. Furthermore, siRNA-mediated IKKβ inhibition reduced xenograft tumor growth and vascularity in vivo. Finally, IKKβ inhibition also affects endothelial cell function in a cancer-independent manner, as IKKβ inhibition reduced pathological retinal angiogenesis in a mouse model of oxygen-induced retinopathy. Taken together, these results provide a novel mechanistic understanding of how the IKKβ pathway affects human lung tumorigenesis, indicating that IKKβ promotes KRAS-induced angiogenesis both by cancer cell-intrinsic and cancer cell-independent mechanisms, which strongly suggests IKKβ inhibition as a promising antiangiogenic approach to be explored for KRAS-induced lung cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Laura B Cardeal
- Chemistry Institute, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Felipe Silva Rodrigues
- Chemistry Institute, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | | | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elena Levantini
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | - Ricardo J Giordano
- Chemistry Institute, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
81
|
Yu M, Sun X, Tyler SR, Liang B, Swatek AM, Lynch TJ, He N, Yuan F, Feng Z, Rotti PG, Choi SH, Shahin W, Liu X, Yan Z, Engelhardt JF. Highly Efficient Transgenesis in Ferrets Using CRISPR/Cas9-Mediated Homology-Independent Insertion at the ROSA26 Locus. Sci Rep 2019; 9:1971. [PMID: 30760763 PMCID: PMC6374392 DOI: 10.1038/s41598-018-37192-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 12/03/2018] [Indexed: 11/09/2022] Open
Abstract
The domestic ferret (Mustela putorius furo) has proven to be a useful species for modeling human genetic and infectious diseases of the lung and brain. However, biomedical research in ferrets has been hindered by the lack of rapid and cost-effective methods for genome engineering. Here, we utilized CRISPR/Cas9-mediated, homology-independent insertion at the ROSA26 "safe harbor" locus in ferret zygotes and created transgenic animals expressing a dual-fluorescent Cre-reporter system flanked by PhiC31 and Bxb1 integrase attP sites. Out of 151 zygotes injected with circular transgene-containing plasmid and Cas9 protein loaded with the ROSA26 intron-1 sgRNA, there were 23 births of which 5 had targeted integration events (22% efficiency). The encoded tdTomato transgene was highly expressed in all tissues evaluated. Targeted integration was verified by PCR analyses, Southern blot, and germ-line transmission. Function of the ROSA26-CAG-LoxPtdTomatoStopLoxPEGFP (ROSA-TG) Cre-reporter was confirmed in primary cells following Cre expression. The Phi31 and Bxb1 integrase attP sites flanking the transgene will also enable rapid directional insertion of any transgene without a size limitation at the ROSA26 locus. These methods and the model generated will greatly enhance biomedical research involving lineage tracing, the evaluation of stem cell therapy, and transgenesis in ferret models of human disease.
Collapse
Affiliation(s)
- Miao Yu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- College of Life Science, Ningxia University, Yinchuan, Ningxia, 750021, China
| | - Xingshen Sun
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Scott R Tyler
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Bo Liang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Anthony M Swatek
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Thomas J Lynch
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Nan He
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Feng Yuan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Zehua Feng
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Pavana G Rotti
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Soon H Choi
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Weam Shahin
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Xiaoming Liu
- College of Life Science, Ningxia University, Yinchuan, Ningxia, 750021, China.
| | - Ziying Yan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
82
|
Han B, Dai Y, Wu H, Zhang Y, Wan L, Zhao J, Liu Y, Xu S, Zhou L. Cimifugin Inhibits Inflammatory Responses of RAW264.7 Cells Induced by Lipopolysaccharide. Med Sci Monit 2019; 25:409-417. [PMID: 30638197 PMCID: PMC6342062 DOI: 10.12659/msm.912042] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND RAW264.7 cells are induced by lipopolysaccharide (LPS) as a rheumatoid arthritis (RA) model. The present study investigated the effect of cimifugin on the proliferation, migration, chemotaxis, and release of inflammation-related factors and inflammation-related signaling pathways of LPS-induced RAW264.7 cells. MATERIAL AND METHODS MTS assay was used to determine the proliferation of RAW264.7 cells. Transwell assay was employed to examine the migration and chemotaxis of the cells. ELISA was performed to measure the contents of chemotactic factors and inflammatory factors in cell culture supernatants. Western blotting was carried out to detect the expression of factors related with MAPKs and NF-κB signaling pathways. RESULTS Cimifugin (0-100 mg/L) had no cytotoxicity for RAW264.7 cells. LPS stimulation induced morphological differentiation of RAW264.7 cells, but intervention by cimifugin inhibited the activation effect by LPS by about 50%. Cimifugin (100 mg/L) decreased the migration and chemotaxis of RAW264.7 cells to 1/3 of that in control cells by decreasing the release of migration- and chemotaxis-associated factors by at least 30%. Cimifugin (100 mg/L) suppressed the release of inflammatory factors from RAW264.7 cells to less than 60% of that in the LPS group. In addition, cimifugin (100 mg/L) inhibited the activities of MAPKs and NF-κB signaling pathways. CONCLUSIONS The present study demonstrates that cimifugin reduces the migration and chemotaxis of RAW264.7 cells and inhibits the release of inflammatory factors and activation of related signaling pathways induced by LPS. Cimifugin may have potential pharmacological effects against RA.
Collapse
Affiliation(s)
- Bin Han
- Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China (mainland).,Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China (mainland).,Department of Pharmacology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Yuan Dai
- Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China (mainland).,Health Rehabilitation Institute, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China (mainland)
| | - Haiyan Wu
- Department of Pharmacology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Yuanyuan Zhang
- Department of Pharmacology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Lihong Wan
- Department of Pharmacology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Jianlei Zhao
- Department of Pharmacology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Yuanqi Liu
- Department of Pharmacology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Shijun Xu
- Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China (mainland).,Health Rehabilitation Institute, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China (mainland)
| | - Liming Zhou
- Department of Pharmacology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| |
Collapse
|
83
|
Nie M, Yang L, Bi X, Wang Y, Sun P, Yang H, Liu P, Li Z, Xia Y, Jiang W. Neutrophil Extracellular Traps Induced by IL8 Promote Diffuse Large B-cell Lymphoma Progression via the TLR9 Signaling. Clin Cancer Res 2018; 25:1867-1879. [PMID: 30446590 DOI: 10.1158/1078-0432.ccr-18-1226] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/21/2018] [Accepted: 11/13/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE More than 30% of patients with diffuse large B-cell lymphoma (DLBCL) experience treatment failure after first-line therapy. Neutrophil extracellular traps (NETs), a pathogen-trapping structure in tumor microenvironment, can promote the transition of autoimmunity to lymphomagenesis. Here, we investigate whether NETs play a novel role in DLBCL progression and its underlying mechanism.Experimental Design: NETs in DLBCL tumor samples and plasma were detected by immunofluorescence and ELISA, respectively. The correlation between NETs and clinical features were analyzed. The effects of NETs on cellular proliferation and migration and mechanisms were explored, and the mechanism of NET formation was also studied by a series of in vitro and in vivo assays. RESULTS Higher levels of NETs in plasma and tumor tissues were associated with dismal outcome in patients with DLBCL. Furthermore, we identified NETs increased cell proliferation and migration in vitro and tumor growth and lymph node dissemination in vivo. Mechanistically, DLBCL-derived IL8 interacted with its receptor (CXCR2) on neutrophils, resulting in the formation of NETs via Src, p38, and ERK signaling. Newly formed NETs directly upregulated the Toll-like receptor 9 (TLR9) pathways in DLBCL and subsequently activated NFκB, STAT3, and p38 pathways to promote tumor progression. More importantly, disruption of NETs, blocking IL8-CXCR2 axis or inhibiting TLR9 could retard tumor progression in preclinical models. CONCLUSIONS Our data reveal a tumor-NETs aggressive interaction in DLBCL and indicate that NETs is a useful prognostic biomarker and targeting this novel cross-talk represents a new therapeutic opportunity in this challenging disease.
Collapse
Affiliation(s)
- Man Nie
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, P.R. China
| | - Linbin Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, P.R. China.,Breast Tumor Center, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, P.R. China
| | - Xiwen Bi
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, P.R. China
| | - Yu Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, P.R. China
| | - Peng Sun
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, P.R. China
| | - Hang Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, P.R. China
| | - Panpan Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, P.R. China
| | - Zhiming Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, P.R. China
| | - Yi Xia
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China. .,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, P.R. China
| | - Wenqi Jiang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China. .,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
84
|
Megakaryocyte Contribution to Bone Marrow Fibrosis: many Arrows in the Quiver. Mediterr J Hematol Infect Dis 2018; 10:e2018068. [PMID: 30416700 PMCID: PMC6223581 DOI: 10.4084/mjhid.2018.068] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/23/2018] [Indexed: 01/14/2023] Open
Abstract
In Primary Myelofibrosis (PMF), megakaryocyte dysplasia/hyperplasia determines the release of inflammatory cytokines that, in turn, stimulate stromal cells and induce bone marrow fibrosis. The pathogenic mechanism and the cells responsible for progression to bone marrow fibrosis in PMF are not completely understood. This review article aims to provide an overview of the crucial role of megakaryocytes in myelofibrosis by discussing the role and the altered secretion of megakaryocyte-derived soluble factors, enzymes and extracellular matrices that are known to induce bone marrow fibrosis.
Collapse
|
85
|
Lierova A, Jelicova M, Nemcova M, Proksova M, Pejchal J, Zarybnicka L, Sinkorova Z. Cytokines and radiation-induced pulmonary injuries. JOURNAL OF RADIATION RESEARCH 2018; 59:709-753. [PMID: 30169853 PMCID: PMC6251431 DOI: 10.1093/jrr/rry067] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/11/2018] [Indexed: 05/20/2023]
Abstract
Radiation therapy is one of the most common treatment strategies for thorax malignancies. One of the considerable limitations of this therapy is its toxicity to normal tissue. The lung is the major dose-limiting organ for radiotherapy. That is because ionizing radiation produces reactive oxygen species that induce lesions, and not only is tumor tissue damaged, but overwhelming inflammatory lung damage can occur in the alveolar epithelium and capillary endothelium. This damage may result in radiation-induced pneumonitis and/or fibrosis. While describing the lung response to irradiation generally, the main focus of this review is on cytokines and their roles and functions within the individual stages. We discuss the relationship between radiation and cytokines and their direct and indirect effects on the formation and development of radiation injuries. Although this topic has been intensively studied and discussed for years, we still do not completely understand the roles of cytokines. Experimental data on cytokine involvement are fragmented across a large number of experimental studies; hence, the need for this review of the current knowledge. Cytokines are considered not only as molecular factors involved in the signaling network in pathological processes, but also for their diagnostic potential. A concentrated effort has been made to identify the significant immune system proteins showing positive correlation between serum levels and tissue damages. Elucidating the correlations between the extent and nature of radiation-induced pulmonary injuries and the levels of one or more key cytokines that initiate and control those damages may improve the efficacy of radiotherapy in cancer treatment and ultimately the well-being of patients.
Collapse
Affiliation(s)
- Anna Lierova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Marcela Jelicova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Marketa Nemcova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Magdalena Proksova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Lenka Zarybnicka
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Zuzana Sinkorova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
- Corresponding author. Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic. Tel.: +420 973 253 219.
| |
Collapse
|
86
|
Lee J, Yoon YJ, Kim JH, Dinh NTH, Go G, Tae S, Park KS, Park HT, Lee C, Roh TY, Di Vizio D, Gho YS. Outer Membrane Vesicles Derived From Escherichia coli Regulate Neutrophil Migration by Induction of Endothelial IL-8. Front Microbiol 2018; 9:2268. [PMID: 30369908 PMCID: PMC6194319 DOI: 10.3389/fmicb.2018.02268] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 09/05/2018] [Indexed: 12/14/2022] Open
Abstract
Outer membrane vesicles (OMVs) are spherical, proteolipid nanostructures that are constitutively released by Gram-negative bacteria including Escherichia coli. Although it has been shown that administration of E. coli OMVs stimulates a strong pulmonary inflammatory response with infiltration of neutrophils into the lungs in vivo, the mechanism of E. coli OMV-mediated neutrophil recruitment is poorly characterized. In this study, we observed significant infiltration of neutrophils into the mouse lung tissues in vivo, with increased expression of the neutrophil chemoattractant CXCL1, a murine functional homolog of human IL-8, on intraperitoneal administration of E. coli OMVs. In addition, OMVs and CD31-positive endothelial cells colocalized in the mouse lungs. Moreover, in vitro results showed that E. coli OMVs significantly increased IL-8 release from human microvascular endothelial cells and toll-like receptor (TLR)4 was found to be the main component for recognizing E. coli OMVs among human endothelial cell-associated TLRs. Furthermore, the transmigration of neutrophils was suppressed in the lung tissues obtained from TLR4 knockout mice treated with E. coli OMVs. Taken together, our data demonstrated that E. coli OMVs potently recruit neutrophils into the lung via the release of IL-8/CXCL1 from endothelial cells in TLR4- and NF-κB-dependent manners.
Collapse
Affiliation(s)
- Jaewook Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Yae Jin Yoon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Ji Hyun Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Nhung Thi Hong Dinh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Gyeongyun Go
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Sookil Tae
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Kyong-Su Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Hyun Taek Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Changjin Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Tae-Young Roh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Dolores Di Vizio
- Division of Cancer Biology and Therapeutics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| |
Collapse
|
87
|
Coburn LA, Singh K, Asim M, Barry DP, Allaman MM, Al-Greene NT, Hardbower DM, Polosukhina D, Williams CS, Delgado AG, Piazuelo MB, Washington MK, Gobert AP, Wilson KT. Loss of solute carrier family 7 member 2 exacerbates inflammation-associated colon tumorigenesis. Oncogene 2018; 38:1067-1079. [PMID: 30202097 PMCID: PMC6377304 DOI: 10.1038/s41388-018-0492-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/19/2018] [Accepted: 08/17/2018] [Indexed: 01/06/2023]
Abstract
Solute carrier family 7 member 2 (SLC7A2, also known as CAT2) is an inducible transporter of the semi-essential amino acid L-arginine (L-Arg), which has been implicated in wound repair. We have reported that both SLC7A2 expression and L-Arg availability are decreased in colonic tissues from inflammatory bowel disease patients and that mice lacking Slc7a2 exhibit a more severe disease course when exposed to dextran sulfate sodium (DSS) compared to wild-type (WT) mice. Here, we present evidence that SLC7A2 plays a role in modulating colon tumorigenesis in the azoxymethane(AOM)-DSS model of colitis-associated carcinogenesis (CAC). SLC7A2 was localized predominantly to colonic epithelial cells in WT mice. Utilizing the AOM-DSS model, Slc7a2–/– mice had significantly increased tumor number, burden, and risk of high-grade dysplasia versus WT mice. Tumors from Slc7a2–/– mice exhibited significantly increased levels of the proinflammatory cytokines/chemokines IL-1β, CXCL1, CXCL5, IL-3, CXCL2, CCL3, and CCL4, but decreased levels of IL-4, CXCL9, and CXCL10 compared to tumors from WT mice. This was accompanied by a shift toward pro-tumorigenic M2 macrophage activation in Slc7a2-deficient mice, as marked by increased colonic CD11b+F4/80+ARG1+ cells with no alteration in CD11b+F4/80+NOS2+ cells by flow cytometry and immunofluorescence microscopy. The shift toward M2 macrophage activation was confirmed in bone marrow-derived macrophages from Slc7a2–/– mice. In bone marrow chimeras between Slc7a2–/– and WT mice, the recipient genotype drove the CAC phenotype, suggesting the importance of epithelial SLC7A2 in abrogating neoplastic risk. These data reveal that SLC7A2 has a significant role in the protection from CAC in the setting of chronic colitis, and suggest that the decreased SLC7A2 in inflammatory bowel disease (IBD) may contribute to CAC risk. Strategies to enhance L-Arg availability by supplementing L-Arg and/or increasing L-Arg uptake could represent a therapeutic approach in IBD to reduce the substantial long-term risk of colorectal carcinoma.
Collapse
Affiliation(s)
- Lori A Coburn
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA.,Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kshipra Singh
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel P Barry
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Margaret M Allaman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicole T Al-Greene
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dana M Hardbower
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dina Polosukhina
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher S Williams
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA.,Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alberto G Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alain P Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T Wilson
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA. .,Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA. .,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA. .,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA. .,Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
88
|
Tan S, Wang K, Sun F, Li Y, Gao Y. CXCL9 promotes prostate cancer progression through inhibition of cytokines from T cells. Mol Med Rep 2018; 18:1305-1310. [PMID: 29901197 PMCID: PMC6072144 DOI: 10.3892/mmr.2018.9152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 03/27/2018] [Indexed: 01/29/2023] Open
Abstract
Chemokines have been demonstrated to serve an important role in a variety of diseases, particularly in tumor progression. There have been numerous studies that have reported that T cells serve major roles in tumor progression. However, the function of CXC motif chemokine ligand 9 (CXCL9) in prostate cancer remains unknown. The present study aimed to investigate the role of CXCL9 in prostate cancer. A prostate cancer mouse model was generated by treating C57/BL‑6 and B6.Cg‑Selplgtm1Fur/J mice with 3,2'‑dimethyl 4‑aminobiphenyl (DMAB). Hematoxylin and eosin staining detected the histopathological alterations of mouse prostate tissues. Immunohistochemistry (IHC) staining determined cell proliferation of the mice. Flow cytometry was used to detect the alterations of T cells in C57+DMAB or CXCL9+DMAB mice. Immunofluorescence revealed that there was positive expression of interleukin‑6 (IL‑6) and transforming growth factor (TGF)‑β in the mouse tissues. The survival rates of C57+DMAB and CXCL9+DMAB mice was analyzed. The association of CXCL9 expression and clinical stages was also evaluated. Results revealed that prostate cancer pathology and cell proliferation in CXCL9+DMAB mice were significantly greater compared with the C57+DMAB mice. Compared with C57+DMAB mice, the number of T cells in peripheral blood and spleen of CXCL9+DMAB mice was significantly reduced. IHC demonstrated that the expression of IL‑6 and TGF‑β was significantly downregulated in the CXCL9+DMAB mice. The survival rate of CXCL9+DMAB mice was significantly decreased compared with the C57+DMAB mice. In addition, reverse transcription‑quantitative polymerase chain reaction analysis demonstrated that CXCL9 mRNA expression in clinical samples was positively associated with clinical pathological stages of prostate cancer. In conclusion, CXCL9 may promote prostate cancer progression via inhibition of cytokines from T cells.
Collapse
Affiliation(s)
- Shanfeng Tan
- Department of Urology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Kai Wang
- Department of Urology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Fuguang Sun
- Department of Urology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Yang Li
- Department of Urology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Yisheng Gao
- Department of Urology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
89
|
De Cunto G, Bartalesi B, Cavarra E, Balzano E, Lungarella G, Lucattelli M. Ongoing Lung Inflammation and Disease Progression in Mice after Smoking Cessation: Beneficial Effects of Formyl-Peptide Receptor Blockade. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2195-2206. [PMID: 30031729 DOI: 10.1016/j.ajpath.2018.06.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/04/2018] [Accepted: 06/18/2018] [Indexed: 01/24/2023]
Abstract
The most important risk factor for chronic obstructive pulmonary disease (COPD) is cigarette smoking. Until now, smoking cessation (SC) is the only treatment effective in slowing down the progression of the disease. However, in many cases SC may only relieve the airflow obstruction and inflammatory response. Consequently, a persistent lung inflammation in ex-smokers is associated with progressive deterioration of respiratory functions. This is an increasingly important clinical problem whose mechanistic basis remains poorly understood. Available therapies do not adequately suppress inflammation and are not able to stop the vicious cycle that is at the basis of persistent inflammation. In addition, in mice after SC an ongoing inflammation and progressive lung deterioration is observed. After 4 months of smoke exposure mice show mild emphysematous changes. Lung inflammation is still present after SC, and emphysema progresses during the next 6-month period of observation. Destruction of alveolar walls is associated with airways remodeling (goblet cell metaplasia and peribronchiolar fibrosis). Modulation of formyl-peptide receptor signaling with antagonists mitigates inflammation and prevents deterioration of lung structures. This study suggests an important role for N-formylated peptides in the progression and exacerbation of COPD. Modulating formyl-peptide receptor signal should be explored as a potential new therapy for COPD.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emilia Balzano
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.
| |
Collapse
|
90
|
Jin SP, Li Z, Choi EK, Lee S, Kim YK, Seo EY, Chung JH, Cho S. Urban particulate matter in air pollution penetrates into the barrier-disrupted skin and produces ROS-dependent cutaneous inflammatory response in vivo. J Dermatol Sci 2018; 91:S0923-1811(18)30202-0. [PMID: 29731195 DOI: 10.1016/j.jdermsci.2018.04.015] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/22/2018] [Accepted: 04/23/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Particulate matter (PM) is an integral part of air pollution, which is a mixture of particles suspended in the air. Recently, it has been reported that PM is associated with increased risks of skin diseases, especially atopic dermatitis in children. However, it is unclear if PM directly goes into the skin and what mechanisms are involved in response to PM. OBJECTIVE To see whether PM could penetrate into the barrier-disrupted skin, produce reactive oxygen species (ROS), and elicit an inflammatory response. METHODS We collected PMs during a winter in Seoul and used cultured keratinocytes for in vitro study and tape-stripped BALB/c mice for in vivo study. RESULTS Keratinocyte cytotoxicity increased in a dose-dependent manner by PM treatment. IL-8 and MMP-1 mRNA expression and protein levels were significantly increased compared to control by qPCR and ELISA, respectively. Cellular ROS production was increased by PM treatment, and antioxidant N-acetyl cysteine pretreatment prevented induction of inflammatory cytokines IL-8 and MMP-1. In PM-treated keratinocytes, electron-dense subcellular particles were observed by transmission electron microscopy. PM was observed inside hair follicles in both intact and barrier-disrupted skin in vivo. Additionally, intercellular penetration of PM was seen in the barrier-disrupted skin. Repeated PM application induced epidermal thickening and dermal inflammation with neutrophil infiltration. Finally, N-acetyl cysteine could ameliorate skin inflammation induced by PM application. CONCLUSION PM penetrates into the barrier-disrupted skin, causing inflammation, demonstrating detrimental effects in the skin.
Collapse
Affiliation(s)
- Seon-Pil Jin
- Department of Dermatology, Seoul National University College of Medicine, Republic of Korea; Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Republic of Korea; Deparment of Biomedical Science, Seoul National University Graduate School, Republic of Korea
| | - Zhenyu Li
- Department of Dermatology, Seoul National University College of Medicine, Republic of Korea; Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Republic of Korea; Deparment of Biomedical Science, Seoul National University Graduate School, Republic of Korea
| | - Eun Kyung Choi
- Laboratory of Electron Microscope, Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Serah Lee
- Department of Dermatology, Seoul National University College of Medicine, Republic of Korea; Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Republic of Korea
| | - Yoen Kyung Kim
- Department of Dermatology, Seoul National University College of Medicine, Republic of Korea; Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Republic of Korea
| | - Eun Young Seo
- Department of Dermatology, Seoul National University College of Medicine, Republic of Korea; Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, Republic of Korea; Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Republic of Korea; Deparment of Biomedical Science, Seoul National University Graduate School, Republic of Korea
| | - Soyun Cho
- Department of Dermatology, Seoul National University College of Medicine, Republic of Korea; Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Republic of Korea; Department of Dermatology, Seoul National University Boramae Medical Center, Republic of Korea.
| |
Collapse
|
91
|
Taipale K, Tähtinen S, Havunen R, Koski A, Liikanen I, Pakarinen P, Koivisto-Korander R, Kankainen M, Joensuu T, Kanerva A, Hemminki A. Interleukin 8 activity influences the efficacy of adenoviral oncolytic immunotherapy in cancer patients. Oncotarget 2018; 9:6320-6335. [PMID: 29464075 PMCID: PMC5814215 DOI: 10.18632/oncotarget.23967] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 12/12/2017] [Indexed: 12/20/2022] Open
Abstract
After the landmark approval of T-VEC, oncolytic viruses are finding their way to the clinics. However, response rates have still room for improvement, and unfortunately there are currently no available markers to predict responses for oncolytic immunotherapy. Interleukin 8 (IL-8) production is upregulated in many cancers and it also connects to several pathways that have been shown to impair the efficacy of adenoviral immunotherapy. We studied the role of IL-8 in 103 cancer patients treated with oncolytic adenoviruses. We found high baseline serum IL-8 concentration to be independently associated with poor prognosis (p<0.001). Further, normal baseline IL-8 was associated with improved prognostic potential of calculation of the neutrophil-to-lymphocyte ratio (p<0.001). Interestingly, a decrease in IL-8 concentration after treatment with oncolytic adenovirus predicted better overall survival (p<0.001) and higher response rate, although this difference was not significant (p=0.066). We studied the combination of adenovirus and IL-8 neutralizing antibody ex vivo in single cell suspensions and in co-cultures of tumor-associated CD15+ neutrophils and CD3+ tumor-infiltrating lymphocytes derived from fresh patient tumor samples. These results indicate a role for IL-8 as a biomarker in oncolytic virotherapy, but additionally provide a rationale for targeting IL-8 to improve treatment efficacy. In conclusion, curtailing the activity of IL-8 systemically or locally in the tumor microenvironment could improve anti-tumor immune responses resulting in enhanced efficacy of adenoviral immunotherapy of cancer.
Collapse
Affiliation(s)
- Kristian Taipale
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| | - Siri Tähtinen
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| | - Anniina Koski
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland.,Department of Neurosurgery, HUCH, Helsinki, Finland
| | - Ilkka Liikanen
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| | - Päivi Pakarinen
- Department of Obstetrics and Gynecology, HUCH, Helsinki, Finland
| | | | - Matti Kankainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | | | - Anna Kanerva
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland.,Department of Obstetrics and Gynecology, HUCH, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland.,Docrates Cancer Center, Helsinki, Finland.,TILT Biotherapeutics Ltd., Helsinki, Finland.,Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
92
|
The Systemic Response to Topical Aldara Treatment is Mediated Through Direct TLR7 Stimulation as Imiquimod Enters the Circulation. Sci Rep 2017; 7:16570. [PMID: 29185473 PMCID: PMC5707416 DOI: 10.1038/s41598-017-16707-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/08/2017] [Indexed: 12/13/2022] Open
Abstract
Topical application of Aldara cream, containing the Toll-like receptor 7/8 agonist Imiquimod, is a widely used mouse model for investigating the pathogenesis of psoriasis. We have previously used this model to study the effects of peripheral inflammation on the brain, and reported a brain-specific response characterised by increased transcription, infiltration of immune cells and anhedonic-like behavior. Here, we perform a more robust characterisation of the systemic response to Aldara application and find a potent but transient response in the periphery, followed by a prolonged response in the brain. Mass spectrometry analysis of plasma and brain samples identified significant levels of Imiquimod in both compartments at molar concentrations likely to evoke a biological response. Indeed, the association of Imiquimod with the brain correlated with increased Iba1 and GFAP staining, indicative of microglia and astrocyte reactivity. These results highlight the potency of this model and raise the question of how useful it is for interpreting the systemic response in psoriasis-like skin inflammation. In addition, the potential impact on the brain should be considered with regards to human use and may explain why fatigue, headaches and nervousness have been reported as side effects following prolonged Aldara use.
Collapse
|
93
|
Roca H, Jones JD, Purica MC, Weidner S, Koh AJ, Kuo R, Wilkinson JE, Wang Y, Daignault-Newton S, Pienta KJ, Morgan TM, Keller ET, Nör JE, Shea LD, McCauley LK. Apoptosis-induced CXCL5 accelerates inflammation and growth of prostate tumor metastases in bone. J Clin Invest 2017; 128:248-266. [PMID: 29202471 DOI: 10.1172/jci92466] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 10/17/2017] [Indexed: 12/31/2022] Open
Abstract
During tumor progression, immune system phagocytes continually clear apoptotic cancer cells in a process known as efferocytosis. However, the impact of efferocytosis in metastatic tumor growth is unknown. In this study, we observed that macrophage-driven efferocytosis of prostate cancer cells in vitro induced the expression of proinflammatory cytokines such as CXCL5 by activating Stat3 and NF-κB(p65) signaling. Administration of a dimerizer ligand (AP20187) triggered apoptosis in 2 in vivo syngeneic models of bone tumor growth in which apoptosis-inducible prostate cancer cells were either coimplanted with vertebral bodies, or inoculated in the tibiae of immunocompetent mice. Induction of 2 pulses of apoptosis correlated with increased infiltration of inflammatory cells and accelerated tumor growth in the bone. Apoptosis-induced tumors displayed elevated expression of the proinflammatory cytokine CXCL5. Likewise, CXCL5-deficient mice had reduced tumor progression. Peripheral blood monocytes isolated from patients with bone metastasis of prostate cancer were more efferocytic compared with normal controls, and CXCL5 serum levels were higher in metastatic prostate cancer patients relative to patients with localized prostate cancer or controls. Altogether, these findings suggest that the myeloid phagocytic clearance of apoptotic cancer cells accelerates CXCL5-mediated inflammation and tumor growth in bone, pointing to CXCL5 as a potential target for cancer therapeutics.
Collapse
Affiliation(s)
- Hernan Roca
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Jacqueline D Jones
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Marta C Purica
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Savannah Weidner
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Amy J Koh
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Robert Kuo
- Department of Chemical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - John E Wilkinson
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yugang Wang
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Stephanie Daignault-Newton
- Department of Biostatistics, Center for Cancer Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Kenneth J Pienta
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Todd M Morgan
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Evan T Keller
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Jacques E Nör
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - Laurie K McCauley
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
94
|
Cagle LA, Franzi LM, Linderholm AL, Last JA, Adams JY, Harper RW, Kenyon NJ. Effects of positive end-expiratory pressure and recruitment maneuvers in a ventilator-induced injury mouse model. PLoS One 2017; 12:e0187419. [PMID: 29112971 PMCID: PMC5675408 DOI: 10.1371/journal.pone.0187419] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/19/2017] [Indexed: 12/26/2022] Open
Abstract
Background Positive-pressure mechanical ventilation is an essential therapeutic intervention, yet it causes the clinical syndrome known as ventilator-induced lung injury. Various lung protective mechanical ventilation strategies have attempted to reduce or prevent ventilator-induced lung injury but few modalities have proven effective. A model that isolates the contribution of mechanical ventilation on the development of acute lung injury is needed to better understand biologic mechanisms that lead to ventilator-induced lung injury. Objectives To evaluate the effects of positive end-expiratory pressure and recruitment maneuvers in reducing lung injury in a ventilator-induced lung injury murine model in short- and longer-term ventilation. Methods 5–12 week-old female BALB/c mice (n = 85) were anesthetized, placed on mechanical ventilation for either 2 hrs or 4 hrs with either low tidal volume (8 ml/kg) or high tidal volume (15 ml/kg) with or without positive end-expiratory pressure and recruitment maneuvers. Results Alteration of the alveolar-capillary barrier was noted at 2 hrs of high tidal volume ventilation. Standardized histology scores, influx of bronchoalveolar lavage albumin, proinflammatory cytokines, and absolute neutrophils were significantly higher in the high-tidal volume ventilation group at 4 hours of ventilation. Application of positive end-expiratory pressure resulted in significantly decreased standardized histology scores and bronchoalveolar absolute neutrophil counts at low- and high-tidal volume ventilation, respectively. Recruitment maneuvers were essential to maintain pulmonary compliance at both 2 and 4 hrs of ventilation. Conclusions Signs of ventilator-induced lung injury are evident soon after high tidal volume ventilation (as early as 2 hours) and lung injury worsens with longer-term ventilation (4 hrs). Application of positive end-expiratory pressure and recruitment maneuvers are protective against worsening VILI across all time points. Dynamic compliance can be used guide the frequency of recruitment maneuvers to help ameloriate ventilator-induced lung injury.
Collapse
Affiliation(s)
- Laura A. Cagle
- Center for Comparative Respiratory Biology and Medicine, University of California, Davis, Davis, CA, United States of America
- * E-mail:
| | - Lisa M. Franzi
- Center for Comparative Respiratory Biology and Medicine, University of California, Davis, Davis, CA, United States of America
| | - Angela L. Linderholm
- Center for Comparative Respiratory Biology and Medicine, University of California, Davis, Davis, CA, United States of America
| | - Jerold A. Last
- Center for Comparative Respiratory Biology and Medicine, University of California, Davis, Davis, CA, United States of America
| | - Jason Y. Adams
- Division of Pulmonary, Critical Care, and Sleep Medicine, School of Medicine, University of California, Davis, Davis, CA, United States of America
| | - Richart W. Harper
- Center for Comparative Respiratory Biology and Medicine, University of California, Davis, Davis, CA, United States of America
- Division of Pulmonary, Critical Care, and Sleep Medicine, School of Medicine, University of California, Davis, Davis, CA, United States of America
| | - Nicholas J. Kenyon
- Center for Comparative Respiratory Biology and Medicine, University of California, Davis, Davis, CA, United States of America
- Division of Pulmonary, Critical Care, and Sleep Medicine, School of Medicine, University of California, Davis, Davis, CA, United States of America
| |
Collapse
|
95
|
Dong G, Song L, Tian C, Wang Y, Miao F, Zheng J, Lu C, Alsadun S, Graves DT. FOXO1 Regulates Bacteria-Induced Neutrophil Activity. Front Immunol 2017; 8:1088. [PMID: 28928749 PMCID: PMC5591501 DOI: 10.3389/fimmu.2017.01088] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/21/2017] [Indexed: 02/05/2023] Open
Abstract
Neutrophils play an essential role in the innate immune response to microbial infection and are particularly important in clearing bacterial infection. We investigated the role of the transcription factor FOXO1 in the response of neutrophils to bacterial challenge with Porphyromonas gingivalis in vivo and in vitro. In these experiments, the effect of lineage-specific FOXO1 deletion in LyzM.Cre+FOXO1L/L mice was compared with matched littermate controls. FOXO1 deletion negatively affected several critical aspects of neutrophil function in vivo including mobilization of neutrophils from the bone marrow (BM) to the vasculature, recruitment of neutrophils to sites of bacterial inoculation, and clearance of bacteria. In vitro FOXO1 regulated neutrophil chemotaxis and bacterial killing. Moreover, bacteria-induced expression of CXCR2 and CD11b, which are essential for several aspects of neutrophil function, was dependent on FOXO1 in vivo and in vitro. Furthermore, FOXO1 directly interacted with the promoter regions of CXCR2 and CD11b. Bacteria-induced nuclear localization of FOXO1 was dependent upon toll-like receptor (TLR) 2 and/or TLR4 and was significantly reduced by inhibitors of reactive oxygen species (ROS and nitric oxide synthase) and deacetylases (Sirt1 and histone deacetylases). These studies show for the first time that FOXO1 activation by bacterial challenge is needed to mobilize neutrophils to transit from the BM to peripheral tissues in response to infection as well as for bacterial clearance in vivo. Moreover, FOXO1 regulates neutrophil function that facilitates chemotaxis, phagocytosis, and bacterial killing.
Collapse
Affiliation(s)
- Guangyu Dong
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liang Song
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Stomatology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Chen Tian
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yu Wang
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Implantology, Stomatology Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fang Miao
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Shanxi Province People's Hospital, Taiyuan, China
| | - Jiabao Zheng
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chanyi Lu
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sarah Alsadun
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
96
|
Dunst J, Kamena F, Matuschewski K. Cytokines and Chemokines in Cerebral Malaria Pathogenesis. Front Cell Infect Microbiol 2017; 7:324. [PMID: 28775960 PMCID: PMC5517394 DOI: 10.3389/fcimb.2017.00324] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022] Open
Abstract
Cerebral malaria is among the major causes of malaria-associated mortality and effective adjunctive therapeutic strategies are currently lacking. Central pathophysiological processes involved in the development of cerebral malaria include an imbalance of pro- and anti-inflammatory responses to Plasmodium infection, endothelial cell activation, and loss of blood-brain barrier integrity. However, the sequence of events, which initiates these pathophysiological processes as well as the contribution of their complex interplay to the development of cerebral malaria remain incompletely understood. Several cytokines and chemokines have repeatedly been associated with cerebral malaria severity. Increased levels of these inflammatory mediators could account for the sequestration of leukocytes in the cerebral microvasculature present during cerebral malaria, thereby contributing to an amplification of local inflammation and promoting cerebral malaria pathogenesis. Herein, we highlight the current knowledge on the contribution of cytokines and chemokines to the pathogenesis of cerebral malaria with particular emphasis on their roles in endothelial activation and leukocyte recruitment, as well as their implication in the progression to blood-brain barrier permeability and neuroinflammation, in both human cerebral malaria and in the murine experimental cerebral malaria model. A better molecular understanding of these processes could provide the basis for evidence-based development of adjunct therapies and the definition of diagnostic markers of disease progression.
Collapse
Affiliation(s)
- Josefine Dunst
- Parasitology Unit, Max Planck Institute for Infection BiologyBerlin, Germany.,Institute of Chemistry and Biochemistry, Free UniversityBerlin, Germany.,Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| | - Faustin Kamena
- Parasitology Unit, Max Planck Institute for Infection BiologyBerlin, Germany.,Institute of Chemistry and Biochemistry, Free UniversityBerlin, Germany.,Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection BiologyBerlin, Germany.,Institute of Chemistry and Biochemistry, Free UniversityBerlin, Germany
| |
Collapse
|
97
|
Hardbower DM, Coburn LA, Asim M, Singh K, Sierra JC, Barry DP, Gobert AP, Piazuelo MB, Washington MK, Wilson KT. EGFR-mediated macrophage activation promotes colitis-associated tumorigenesis. Oncogene 2017; 36:3807-3819. [PMID: 28263971 PMCID: PMC5501754 DOI: 10.1038/onc.2017.23] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 01/12/2017] [Accepted: 01/25/2017] [Indexed: 02/08/2023]
Abstract
Epidermal growth factor receptor (EGFR) signaling is a known mediator of colorectal carcinogenesis. Studies have focused on the role of EGFR signaling in epithelial cells, although the exact nature of the role of EGFR in colorectal carcinogenesis remains a topic of debate. Here, we present evidence that EGFR signaling in myeloid cells, specifically macrophages, is critical for colon tumorigenesis in the azoxymethane-dextran sodium sulfate (AOM-DSS) model of colitis-associated carcinogenesis (CAC). In a human tissue microarray, colonic macrophages demonstrated robust EGFR activation in the pre-cancerous stages of colitis and dysplasia. Utilizing the AOM-DSS model, mice with a myeloid-specific deletion of Egfr had significantly decreased tumor multiplicity and burden, protection from high-grade dysplasia and significantly reduced colitis. Intriguingly, mice with gastrointestinal epithelial cell-specific Egfr deletion demonstrated no differences in tumorigenesis in the AOM-DSS model. The alterations in tumorigenesis in myeloid-specific Egfr knockout mice were accompanied by decreased macrophage, neutrophil and T-cell infiltration. Pro-tumorigenic M2 macrophage activation was diminished in myeloid-specific Egfr-deficient mice, as marked by decreased Arg1 and Il10 mRNA expression and decreased interleukin (IL)-4, IL10 and IL-13 protein levels. Surprisingly, diminished M1 macrophage activation was also detectable, as marked by significantly reduced Nos2 and Il1b mRNA levels and decreased interferon (IFN)-γ, tumor necrosis factor (TNF)-α and IL-1β protein levels. The alterations in M1 and M2 macrophage activation were confirmed in bone marrow-derived macrophages from mice with the myeloid-specific Egfr knockout. The combined effect of restrained M1 and M2 macrophage activation resulted in decreased production of pro-angiogenic factors, CXCL1 and vascular endothelial growth factor (VEGF), and reduced CD31+ blood vessels, which likely contributed to protection from tumorigenesis. These data reveal that EGFR signaling in macrophages, but not in colonic epithelial cells, has a significant role in CAC. EGFR signaling in macrophages may prove to be an effective biomarker of CAC or target for chemoprevention in patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Dana M. Hardbower
- Department of Pathology, Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Lori A. Coburn
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System; Nashville, TN, USA
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Kshipra Singh
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Johanna C. Sierra
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Daniel P. Barry
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Alain P. Gobert
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
| | - M. Blanca Piazuelo
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
| | - M. Kay Washington
- Department of Pathology, Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Keith T. Wilson
- Department of Pathology, Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
- Department of Cancer Biology; Vanderbilt University Medical Center; Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
- Vanderbilt Ingram Cancer Center; Vanderbilt University Medical Center; Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System; Nashville, TN, USA
| |
Collapse
|
98
|
Xue J, Zhao Z, Zhang L, Xue L, Shen S, Wen Y, Wei Z, Wang L, Kong L, Sun H, Ping Q, Mo R, Zhang C. Neutrophil-mediated anticancer drug delivery for suppression of postoperative malignant glioma recurrence. NATURE NANOTECHNOLOGY 2017; 12:692-700. [PMID: 28650441 DOI: 10.1038/nnano.2017.54] [Citation(s) in RCA: 603] [Impact Index Per Article: 86.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/05/2017] [Indexed: 05/19/2023]
Abstract
Cell-mediated drug-delivery systems have received considerable attention for their enhanced therapeutic specificity and efficacy in cancer treatment. Neutrophils (NEs), the most abundant type of immune cells, are known to penetrate inflamed brain tumours. Here we show that NEs carrying liposomes that contain paclitaxel (PTX) can penetrate the brain and suppress the recurrence of glioma in mice whose tumour has been resected surgically. Inflammatory factors released after tumour resection guide the movement of the NEs into the inflamed brain. The highly concentrated inflammatory signals in the brain trigger the release of liposomal PTX from the NEs, which allows delivery of PTX into the remaining invading tumour cells. We show that this NE-mediated delivery of drugs efficiently slows the recurrent growth of tumours, with significantly improved survival rates, but does not completely inhibit the regrowth of tumours.
Collapse
Affiliation(s)
- Jingwei Xue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Zekai Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Shiyang Shen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Yajing Wen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Zhuoyuan Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Lu Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Hongbin Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Qineng Ping
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
99
|
Neutrophil migration in infection and wound repair: going forward in reverse. Nat Rev Immunol 2017; 16:378-91. [PMID: 27231052 DOI: 10.1038/nri.2016.49] [Citation(s) in RCA: 665] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neutrophil migration and its role during inflammation has been the focus of increased interest in the past decade. Advances in live imaging and the use of new model systems have helped to uncover the behaviour of neutrophils in injured and infected tissues. Although neutrophils were considered to be short-lived effector cells that undergo apoptosis in damaged tissues, recent evidence suggests that neutrophil behaviour is more complex and, in some settings, neutrophils might leave sites of tissue injury and migrate back into the vasculature. The role of reverse migration and its contribution to resolution of inflammation remains unclear. In this Review, we discuss the different cues within tissues that mediate neutrophil forward and reverse migration in response to injury or infection and the implications of these mechanisms to human disease.
Collapse
|
100
|
Franck G, Mawson T, Sausen G, Salinas M, Masson GS, Cole A, Beltrami-Moreira M, Chatzizisis Y, Quillard T, Tesmenitsky Y, Shvartz E, Sukhova GK, Swirski FK, Nahrendorf M, Aikawa E, Croce KJ, Libby P. Flow Perturbation Mediates Neutrophil Recruitment and Potentiates Endothelial Injury via TLR2 in Mice: Implications for Superficial Erosion. Circ Res 2017; 121:31-42. [PMID: 28428204 DOI: 10.1161/circresaha.117.310694] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/10/2017] [Accepted: 04/20/2017] [Indexed: 01/25/2023]
Abstract
RATIONALE Superficial erosion currently causes up to a third of acute coronary syndromes; yet, we lack understanding of its mechanisms. Thrombi because of superficial intimal erosion characteristically complicate matrix-rich atheromata in regions of flow perturbation. OBJECTIVE This study tested in vivo the involvement of disturbed flow and of neutrophils, hyaluronan, and Toll-like receptor 2 ligation in superficial intimal injury, a process implicated in superficial erosion. METHODS AND RESULTS In mouse carotid arteries with established intimal lesions tailored to resemble the substrate of human eroded plaques, acute flow perturbation promoted downstream endothelial cell activation, neutrophil accumulation, endothelial cell death and desquamation, and mural thrombosis. Neutrophil loss-of-function limited these findings. Toll-like receptor 2 agonism activated luminal endothelial cells, and deficiency of this innate immune receptor decreased intimal neutrophil adherence in regions of local flow disturbance, reducing endothelial cell injury and local thrombosis (P<0.05). CONCLUSIONS These results implicate flow disturbance, neutrophils, and Toll-like receptor 2 signaling as mechanisms that contribute to superficial erosion, a cause of acute coronary syndrome of likely growing importance in the statin era.
Collapse
Affiliation(s)
- Grégory Franck
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Thomas Mawson
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Grasiele Sausen
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Manuel Salinas
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Gustavo Santos Masson
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Andrew Cole
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Marina Beltrami-Moreira
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Yiannis Chatzizisis
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Thibault Quillard
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Yevgenia Tesmenitsky
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Eugenia Shvartz
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Galina K Sukhova
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Filip K Swirski
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Matthias Nahrendorf
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Elena Aikawa
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Kevin J Croce
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Peter Libby
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.).
| |
Collapse
|