1
|
Long Y, Dai W, Cai K, Xiao Y, Luo A, Lai Z, Wang J, Xu L, Nie H. Systemic Immune Factors and Risk of Allergic Contact Dermatitis: A Bidirectional Mendelian Randomization Study. Int J Mol Sci 2024; 25:10436. [PMID: 39408763 PMCID: PMC11476522 DOI: 10.3390/ijms251910436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/16/2024] [Accepted: 09/21/2024] [Indexed: 10/20/2024] Open
Abstract
Skin inflammation and immune regulation have been suggested to be associated with allergic contact dermatitis (ACD) progression, but whether the system's immune regulation is a cause or a potential mechanism is still unknown. This study aims to assess the upstream and downstream of systemic immune factors on ACD within a bidirectional Mendelian-randomization design. A bidirectional two-sample MR analysis was employed to implement the results from genome-wide association studies for 52 system immune factors and ACD. Genetic associations with systemic immune factors and ACD were obtained from the IEU Open GWAS project database. The inverse-variance weighted (IVW) method was adopted as the primary MR analysis, MR-Egger, weighted median, MR-pleiotropy residual sum, and outlier (MR-PRESSO) was also used as the sensitivity analyses. Only Tumor necrosis factor ligand superfamily member 11 (TNFS11) from among 52 systemic immune factors was associated with a protective effect of ACD. However, ACD was associated with a decrease in Interleukin-9 (IL9) and an increase in C-X-C motif chemokine 1 (GROα), Tumor necrosis factor ligand superfamily member 10 (TRAIL), C4, and complement factor B of the assessed systemic immune factors. This study identified TNFS11 as the upstream regulator and IL9, GROα, TRAIL, C4, and complement factor B as the downstream regulator of ACD, providing opportunities for new therapeutic exploitation of ACD. Nonetheless, these associations of systemic immune factors need to be verified in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lipeng Xu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China; (Y.L.); (W.D.); (K.C.); (Y.X.); (A.L.); (Z.L.); (J.W.)
| | - Hong Nie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China; (Y.L.); (W.D.); (K.C.); (Y.X.); (A.L.); (Z.L.); (J.W.)
| |
Collapse
|
2
|
Kulkarni DH, Starick M, Aponte Alburquerque R, Kulkarni HS. Local complement activation and modulation in mucosal immunity. Mucosal Immunol 2024; 17:739-751. [PMID: 38838816 PMCID: PMC11929374 DOI: 10.1016/j.mucimm.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
The complement system is an evolutionarily conserved arm of innate immunity, which forms one of the first lines of host response to pathogens and assists in the clearance of debris. A deficiency in key activators/amplifiers of the cascade results in recurrent infection, whereas a deficiency in regulating the cascade predisposes to accelerated organ failure, as observed in colitis and transplant rejection. Given that there are over 60 proteins in this system, it has become an attractive target for immunotherapeutics, many of which are United States Food and Drug Administration-approved or in multiple phase 2/3 clinical trials. Moreover, there have been key advances in the last few years in the understanding of how the complement system operates locally in tissues, independent of its activities in circulation. In this review, we will put into perspective the abovementioned discoveries to optimally modulate the spatiotemporal nature of complement activation and regulation at mucosal surfaces.
Collapse
Affiliation(s)
- Devesha H Kulkarni
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marick Starick
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rafael Aponte Alburquerque
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
3
|
Mohammed Y, Tran K, Carlsten C, Ryerson C, Wong A, Lee T, Cheng MP, Vinh DC, Lee TC, Winston BW, Sweet D, Boyd JH, Walley KR, Haljan G, McGeer A, Lamontagne F, Fowler R, Maslove D, Singer J, Patrick DM, Marshall JC, Murthy S, Jain F, Borchers CH, Goodlett DR, Levin A, Russell JA, ARBs CORONA I Consortium. Proteomic Evolution from Acute to Post-COVID-19 Conditions. J Proteome Res 2024; 23:52-70. [PMID: 38048423 PMCID: PMC10775146 DOI: 10.1021/acs.jproteome.3c00324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/30/2023] [Accepted: 11/13/2023] [Indexed: 12/06/2023]
Abstract
Many COVID-19 survivors have post-COVID-19 conditions, and females are at a higher risk. We sought to determine (1) how protein levels change from acute to post-COVID-19 conditions, (2) whether females have a plasma protein signature different from that of males, and (3) which biological pathways are associated with COVID-19 when compared to restrictive lung disease. We measured protein levels in 74 patients on the day of admission and at 3 and 6 months after diagnosis. We determined protein concentrations by multiple reaction monitoring (MRM) using a panel of 269 heavy-labeled peptides. The predicted forced vital capacity (FVC) and diffusing capacity of the lungs for carbon monoxide (DLCO) were measured by routine pulmonary function testing. Proteins associated with six key lipid-related pathways increased from admission to 3 and 6 months; conversely, proteins related to innate immune responses and vasoconstriction-related proteins decreased. Multiple biological functions were regulated differentially between females and males. Concentrations of eight proteins were associated with FVC, %, and they together had c-statistics of 0.751 (CI:0.732-0.779); similarly, concentrations of five proteins had c-statistics of 0.707 (CI:0.676-0.737) for DLCO, %. Lipid biology may drive evolution from acute to post-COVID-19 conditions, while activation of innate immunity and vascular regulation pathways decreased over that period. (ProteomeXchange identifiers: PXD041762, PXD029437).
Collapse
Affiliation(s)
- Yassene Mohammed
- Center
for Proteomics and Metabolomics, Leiden
University Medical Center, Leiden 2333 ZA, The Netherlands
- UVic-Genome
BC Proteomics Centre, University of Victoria, Victoria V8Z 5N3, BC Canada
- Gerald
Bronfman Department of Oncology, McGill
University, Montreal, QC H3A 0G4, Canada
| | - Karen Tran
- Division
of General Internal Medicine, Vancouver
General Hospital and University of British Columbia, 2775 Laurel St, Vancouver, BC V5Z 1M9, Canada
| | - Chris Carlsten
- Division
of Respiratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Christopher Ryerson
- Division
of Respiratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Alyson Wong
- Division
of Respiratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Terry Lee
- Centre for
Health Evaluation and Outcome Science (CHEOS), St. Paul’s Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Matthew P. Cheng
- Division
of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, PQ H4A 3J1, Canada
| | - Donald C. Vinh
- Division
of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, PQ H4A 3J1, Canada
| | - Todd C. Lee
- Division
of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, PQ H4A 3J1, Canada
| | - Brent W. Winston
- Departments
of Critical Care Medicine, Medicine and Biochemistry and Molecular
Biology, Foothills Medical Centre and University
of Calgary, 1403 29 Street
NW, Calgary, Alberta T2N 4N1, Canada
| | - David Sweet
- Division
of Critical Care Medicine, Vancouver General
Hospital, 2775 Laurel St, Vancouver, BC V5Z 1M9, Canada
| | - John H. Boyd
- Centre
for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
- Division of Critical Care Medicine, St.
Paul’s Hospital, University of British
Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Keith R. Walley
- Centre
for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
- Division of Critical Care Medicine, St.
Paul’s Hospital, University of British
Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Greg Haljan
- Department of Medicine, Surrey Memorial
Hospital, 13750 96th
Avenue, Surrey, BC V3V 1Z2, Canada
| | - Allison McGeer
- Mt. Sinai Hospital and University of Toronto, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | | | - Robert Fowler
- Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada
| | - David Maslove
- Department
of Critical Care, Kingston General Hospital
and Queen’s University, 76 Stuart Street, Kingston, ON K7L 2V7, Canada
| | - Joel Singer
- Centre for
Health Evaluation and Outcome Science (CHEOS), St. Paul’s Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - David M. Patrick
- British Columbia Centre for Disease Control
(BCCDC) and University
of British Columbia, 655 West 12th Avenue, Vancouver, BC V5Z 4R4, Canada
| | - John C. Marshall
- Department of Surgery, St. Michael’s
Hospital, 30 Bond Street, Toronto, ON M5B
1W8, Canada
| | - Srinivas Murthy
- BC Children’s Hospital and University of British Columbia, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| | - Fagun Jain
- Black Tusk Research Group, Vancouver, BC V6Z 2C7, Canada
| | - Christoph H. Borchers
- Segal Cancer Proteomics, Centre, Lady Davis
Institute
for Medical Research, McGill University, Montreal, QC H3T 1E2, Canada
- Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Division of Experimental Medicine, McGill
University, Montreal, QC H3T 1E2, Canada
- Department of Pathology, McGill
University, Montreal, QC H3T 1E2, Canada
| | - David R. Goodlett
- UVic-Genome
BC Proteomics Centre, University of Victoria, Victoria V8Z 5N3, BC Canada
| | - Adeera Levin
- Division of Nephrology, St.
Paul’s Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - James A. Russell
- Centre
for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
- Division of Critical Care Medicine, St.
Paul’s Hospital, University of British
Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - ARBs CORONA I Consortium
- Center
for Proteomics and Metabolomics, Leiden
University Medical Center, Leiden 2333 ZA, The Netherlands
- UVic-Genome
BC Proteomics Centre, University of Victoria, Victoria V8Z 5N3, BC Canada
- Gerald
Bronfman Department of Oncology, McGill
University, Montreal, QC H3A 0G4, Canada
- Division
of General Internal Medicine, Vancouver
General Hospital and University of British Columbia, 2775 Laurel St, Vancouver, BC V5Z 1M9, Canada
- Division
of Respiratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Centre for
Health Evaluation and Outcome Science (CHEOS), St. Paul’s Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
- Division
of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, PQ H4A 3J1, Canada
- Departments
of Critical Care Medicine, Medicine and Biochemistry and Molecular
Biology, Foothills Medical Centre and University
of Calgary, 1403 29 Street
NW, Calgary, Alberta T2N 4N1, Canada
- Division
of Critical Care Medicine, Vancouver General
Hospital, 2775 Laurel St, Vancouver, BC V5Z 1M9, Canada
- Centre
for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
- Division of Critical Care Medicine, St.
Paul’s Hospital, University of British
Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, Surrey Memorial
Hospital, 13750 96th
Avenue, Surrey, BC V3V 1Z2, Canada
- Mt. Sinai Hospital and University of Toronto, 600 University Avenue, Toronto, ON M5G 1X5, Canada
- University of Sherbrooke, Sherbrooke, PQ J1K 2R1, Canada
- Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada
- Department
of Critical Care, Kingston General Hospital
and Queen’s University, 76 Stuart Street, Kingston, ON K7L 2V7, Canada
- British Columbia Centre for Disease Control
(BCCDC) and University
of British Columbia, 655 West 12th Avenue, Vancouver, BC V5Z 4R4, Canada
- Department of Surgery, St. Michael’s
Hospital, 30 Bond Street, Toronto, ON M5B
1W8, Canada
- BC Children’s Hospital and University of British Columbia, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
- Black Tusk Research Group, Vancouver, BC V6Z 2C7, Canada
- Segal Cancer Proteomics, Centre, Lady Davis
Institute
for Medical Research, McGill University, Montreal, QC H3T 1E2, Canada
- Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Division of Experimental Medicine, McGill
University, Montreal, QC H3T 1E2, Canada
- Department of Pathology, McGill
University, Montreal, QC H3T 1E2, Canada
- Division of Nephrology, St.
Paul’s Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
4
|
Papadopoulos Z. The role of the cytokine TNF-α in choroidal neovascularization: a systematic review. Eye (Lond) 2024; 38:25-32. [PMID: 37380786 PMCID: PMC10764737 DOI: 10.1038/s41433-023-02634-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 05/06/2023] [Accepted: 06/09/2023] [Indexed: 06/30/2023] Open
Abstract
TNF-α is a multifunctional cytokine produced by macrophages and T cells. This proinflammatory substance is considered to play a crucial role in the inflammatory process associated with age-related macular degeneration (AMD). The current review aimed to describe evidence for an association between TNF-α and AMD reported in various studies. The MEDLINE, Embase, PubMed and Global Health databases were systematically searched to identify studies that investigated the role of TNF-α in AMD. A total of 24 studies were deemed eligible for the review. To better understand and integrate the evidence, the studies were categorised into four major groups in relation to the role of TNF-α in AMD: (1) those examining biological signalling pathways through which TNF-α exerts its effect; (2) investigating levels of TNF-α; (3) exploring the genetics underlying the role of TNF-α; and (4) assessing anti-TNF-α agents as potential treatments for AMD. TNF-α is thought to directly contribute to choroidal neovascularization (CNV) enhancement and has been shown to exert its effect by augmenting the inflammatory response through other signalling pathways. Additionally, different genes have been found to be associated with activities linked to TNF-α in AMD. Overall, measurement of systemic and local levels of TNF-α has not yielded consistent findings, with variable conclusions for the role of anti-TNF-α agents in remission of AMD symptoms. The role of TNF-α in neovascular AMD is not clear, and not all anti-TNF-α agents are safe. The potential of this cytokine in atrophic AMD has not been examined. Future studies should address these unresolved questions.
Collapse
|
5
|
Wang W, Liu S, Zhang S, Zhang J, Tang Y, Zhang W. Incorporating Anticoagulant and Antiplatelet Dual Functional Groups into Thermosetting Polymer Chain for Enhancing Antithrombogenicity. Adv Healthc Mater 2023; 12:e2300680. [PMID: 37515824 DOI: 10.1002/adhm.202300680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/05/2023] [Indexed: 07/31/2023]
Abstract
In clinical practice, high-effective antithrombosis remains a challenge for blood-contacting medical devices. Inspired by the enhanced antithrombogenicity of anticoagulant and antiplatelet combination therapy, a strategy is proposed to synthesize dual-pathway antithrombotic polymers by incorporating anticoagulant and antiplatelet dual functional groups into a single thermosetting polymer chain. The synthesized polymer shows increased antithrombogenicity in vitro, with prolonged activated partial thromboplastin time (APTT) and decreased platelet adhesion. Additionally, it downregulates the expression of coagulation- and inflammation-related factors in rabbit plasma after ex vivo arteriovenous shunt assay and maintains patency of small vascular grafts for at least 6 months without thrombosis on the luminal surface after in vivo replacement of rabbit carotid artery. This work provides a new approach to producing novel antithrombotic polymers for blood-contacting medical devices.
Collapse
Affiliation(s)
- Weizhong Wang
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200240, China
| | - Shaowen Liu
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200240, China
| | - Shan Zhang
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200240, China
| | - Jingjing Zhang
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200240, China
| | - Yuyi Tang
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200240, China
| | - Weijia Zhang
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200240, China
- The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200240, China
- Department of Physiology and Pathophysiology, the Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, School of Basic Medical Sciences, Fudan University, Shanghai, 200240, China
| |
Collapse
|
6
|
Sinkovits G, Schnur J, Hurler L, Kiszel P, Prohászka ZZ, Sík P, Kajdácsi E, Cervenak L, Maráczi V, Dávid M, Zsigmond B, Rimanóczy É, Bereczki C, Willems L, Toonen EJM, Prohászka Z. Evidence, detailed characterization and clinical context of complement activation in acute multisystem inflammatory syndrome in children. Sci Rep 2022; 12:19759. [PMID: 36396679 PMCID: PMC9670087 DOI: 10.1038/s41598-022-23806-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 11/06/2022] [Indexed: 11/18/2022] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a rare, life-threatening complication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. MIS-C develops with high fever, marked inflammation and shock-like picture several weeks after exposure to, or mild infection with SARS-CoV-2. Deep immune profiling identified activated macrophages, neutrophils, B-plasmablasts and CD8 + T cells as key determinants of pathogenesis together with multiple inflammatory markers. The disease rapidly responds to intravenous immunoglobulin (IVIG) treatment with clear changes of immune features. Here we present the results of a comprehensive analysis of the complement system in the context of MIS-C activity and describe characteristic changes during IVIG treatment. We show that activation markers of the classical, alternative and terminal pathways are highly elevated, that the activation is largely independent of anti-SARS-CoV-2 humoral immune response, but is strongly associated with markers of macrophage activation. Decrease of complement activation is closely associated with rapid improvement of MIS-C after IVIG treatment.
Collapse
Affiliation(s)
- György Sinkovits
- grid.11804.3c0000 0001 0942 9821Department of Internal Medicine and Hematology, Semmelweis University, Budapest, 1085 Hungary
| | - János Schnur
- grid.413987.00000 0004 0573 5145Heim Pál National Pediatric Institute, Budapest, 1089 Hungary
| | - Lisa Hurler
- grid.11804.3c0000 0001 0942 9821Department of Internal Medicine and Hematology, Semmelweis University, Budapest, 1085 Hungary
| | - Petra Kiszel
- grid.11804.3c0000 0001 0942 9821Research Group for Immunology and Hematology, Semmelweis University-Eötvös Loránd Research Network (Office for Supported Research Groups), Budapest, 1085 Hungary
| | - Zita Z. Prohászka
- grid.11804.3c0000 0001 0942 9821Department of Internal Medicine and Hematology, Semmelweis University, Budapest, 1085 Hungary
| | - Pál Sík
- grid.11804.3c0000 0001 0942 9821Department of Internal Medicine and Hematology, Semmelweis University, Budapest, 1085 Hungary
| | - Erika Kajdácsi
- grid.11804.3c0000 0001 0942 9821Department of Internal Medicine and Hematology, Semmelweis University, Budapest, 1085 Hungary
| | - László Cervenak
- grid.11804.3c0000 0001 0942 9821Department of Internal Medicine and Hematology, Semmelweis University, Budapest, 1085 Hungary
| | - Veronika Maráczi
- grid.413987.00000 0004 0573 5145Heim Pál National Pediatric Institute, Budapest, 1089 Hungary
| | - Máté Dávid
- grid.413987.00000 0004 0573 5145Heim Pál National Pediatric Institute, Budapest, 1089 Hungary
| | - Borbála Zsigmond
- grid.413987.00000 0004 0573 5145Heim Pál National Pediatric Institute, Budapest, 1089 Hungary
| | - Éva Rimanóczy
- grid.413987.00000 0004 0573 5145Heim Pál National Pediatric Institute, Budapest, 1089 Hungary
| | - Csaba Bereczki
- grid.9008.10000 0001 1016 9625Department of Pediatrics, University of Szeged, Szeged, 6720 Hungary
| | - Loek Willems
- grid.435189.2R&D Department, Hycult Biotech, 5405 PB Uden, The Netherlands
| | - Erik J. M. Toonen
- grid.435189.2R&D Department, Hycult Biotech, 5405 PB Uden, The Netherlands
| | - Zoltán Prohászka
- grid.11804.3c0000 0001 0942 9821Department of Internal Medicine and Hematology, Semmelweis University, Budapest, 1085 Hungary ,grid.11804.3c0000 0001 0942 9821Research Group for Immunology and Hematology, Semmelweis University-Eötvös Loránd Research Network (Office for Supported Research Groups), Budapest, 1085 Hungary
| |
Collapse
|
7
|
Relationship between Plasma Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Level and Proteome Profile of Cows. Animals (Basel) 2022; 12:ani12121559. [PMID: 35739894 PMCID: PMC9219453 DOI: 10.3390/ani12121559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/02/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic and multifunctional neuropeptide; it takes part in the regulation of various physiological processes, such as feeding, reproduction, catecholamine synthesis, thermoregulation, motor activity, brain development and neuronal survival. Since PACAP plays important regulatory roles, we hypothesized that the level of PACAP in blood is associated with expression of other proteins, which are involved in different metabolic pathways. The objective of the present study was to compare plasma protein profiles of cows with high and low plasma PACAP levels. Differential proteome analyses were performed by two-dimensional gel electrophoresis (2D-PAGE) followed by tryptic digestion and protein identification by liquid chromatography−mass spectrometry (LC-MS). A total of 210 protein spots were detected, and 16 protein spots showed statistically significant differences (p < 0.05) in the expression levels between groups. Ten spots showed a higher intensity in the high-PACAP-concentration group, while six spots were more abundant in the low-PACAP-concentration group. The functions of the differentially expressed proteins indicate that the PACAP level of plasma is related to the lipid metabolism and immune status of cattle.
Collapse
|
8
|
Lee MJ, Na K, Shin H, Kim CY, Cho JY, Kang CM, Kim SH, Kim H, Choi HJ, Lee CK, Bae S, Son S, Paik YK. Early Diagnostic Ability of Human Complement Factor B in Pancreatic Cancer Is Partly Linked to Its Potential Tumor-Promoting Role. J Proteome Res 2021; 20:5315-5328. [PMID: 34766501 DOI: 10.1021/acs.jproteome.1c00805] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although plasma complement factor B (CFB, NX_P00751), both alone and in combination with CA19-9 (i.e., the ComB-CAN), previously exhibited a reliable diagnostic ability for pancreatic cancer (PC), its detectability of the early stages and the cancer detection mechanism remained elusive. We first evaluated the diagnostic accuracy of ComB-CAN using plasma samples from healthy donors (HDs), patients with chronic pancreatitis (CP), and patients with different PC stages (I/II vs III/IV). An analysis of the area under the curve (AUC) by PanelComposer using logistic regression revealed that ComB-CAN has a superior diagnostic ability for early-stage PC (97.1.% [95% confidence interval (CI): (97.1-97.2)]) compared with CFB (94.3% [95% CI: 94.2-94.4]) or CA19-9 alone (34.3% [95% CI: 34.1-34.4]). In the comparisons of all stages of patients with PC vs CP and HDs, the AUC values of ComB-CAN, CFB, and CA19-9 were 0.983 (95% CI: 0.983-0.983), 0.950 (95% CI: 0.950-0.951), and 0.873 (95% CI: 0.873-0.874), respectively. We then investigated the molecular mechanism underlying the detection of early-stage PC by using stable cell lines of CFB knockdown and CFB overexpression. A global transcriptomic analysis coupled to cell invasion assays of both CFB-modulated cell lines suggested that CFB plays a tumor-promoting role in PC, which likely initiates the PI3K-AKT cancer signaling pathway. Thus our study establishes ComB-CAN as a reliable early diagnostic marker for PC that can be clinically applied for early PC screening in the general public.
Collapse
Affiliation(s)
- Min Jung Lee
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Keun Na
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Heon Shin
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Chae-Yeon Kim
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Jin-Young Cho
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | | | | | | | | | | | - Sumi Bae
- JW Bioscience Corp., 2477, Nambusunhwan-ro, Seocho-gu, Seoul 06725, South Korea
| | - Sunghwa Son
- JW Holdings Corp., 2477, Nambusunhwan-ro, Seocho-gu, Seoul 06725, South Korea
| | - Young-Ki Paik
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| |
Collapse
|
9
|
Li X, Zhang W, Li P, Lu G. The protective effect and mechanism of lentinan on acute kidney injury in septic rats. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:883. [PMID: 32793727 DOI: 10.21037/atm-20-5158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND This study aimed to investigate the protective effect and mechanism of lentinan (LNT) on acute kidney injury (AKI) in septic rats. METHODS A total 72 male SD rats were randomly divided into 6 groups with 12 rats in each group. Except for the sham group, all groups, including the burn sepsis group (BS group), the positive drug control group (dexamethasone, 5 mg/kg, PC group), the LNT low-concentration group (LNT-L group) (50 mg/kg), the LNT medium-concentration group (LNT-M group) (100 mg/kg), and the LNT high-concentration group (LNT-H group) (200 mg/kg), were intraperitoneally injected with the same amount of normal saline 30 min before injury. The levels of serum interleukin (IL)-4, IL-6, IL-10, and tumor necrosis factor alpha (TNF-α); the indexes of blood urea nitrogen (BUN) and creatinine (Cr); and the protein expression levels of inducible nitric oxide synthase (iNOS), intercellular adhesion molecule 1 (ICAM-1), and nuclear factor-κB (NF-κB) in renal tissue were detected 24 hours after the model was established. RESULTS Compared with the sham group, the BUN and Cr of the other groups were significantly higher, while those of the LNT group with different concentrations were significantly lower than those of the BS group (P<0.05). Compared with the sham group, the protein expression levels of NF-κB, iNOS, and ICAM-1 along with the levels of pro-inflammatory factors TNF-α and IL-6 in serum were significantly increased, while the levels of anti-inflammatory factors IL-4 and IL-10 were obviously lower in the BS group. Compared with the BS group, the protein expression levels of NF-κB, iNOS, and ICAM-1 along with the levels of pro-inflammatory factors TNF-α and IL-6 in serum were significantly decreased, while the levels of anti-inflammatory factors IL-4 and IL-10 were obviously increased in the LNT group with different concentrations.. CONCLUSIONS LNT has a certain protective effect on AKI in septic rats, and its mechanism may involve inhibiting the activation of NF-κB, which suppresses the expression of proinflammatory factors in turn, thus promoting the release of anti-inflammatory factors.
Collapse
Affiliation(s)
- Xiaoping Li
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Nephrology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Wanfen Zhang
- Department of Nephrology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Ping Li
- Department of Nephrology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Guoyuan Lu
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
10
|
Mola S, Foisy S, Boucher G, Major F, Beauchamp C, Karaky M, Goyette P, Lesage S, Rioux JD. A transcriptome-based approach to identify functional modules within and across primary human immune cells. PLoS One 2020; 15:e0233543. [PMID: 32469933 PMCID: PMC7259617 DOI: 10.1371/journal.pone.0233543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 05/07/2020] [Indexed: 11/20/2022] Open
Abstract
Genome-wide transcriptomic analyses have provided valuable insight into fundamental biology and disease pathophysiology. Many studies have taken advantage of the correlation in the expression patterns of the transcriptome to infer a potential biologic function of uncharacterized genes, and multiple groups have examined the relationship between co-expression, co-regulation, and gene function on a broader scale. Given the unique characteristics of immune cells circulating in the blood, we were interested in determining whether it was possible to identify functional co-expression modules in human immune cells. Specifically, we sequenced the transcriptome of nine immune cell types from peripheral blood cells of healthy donors and, using a combination of global and targeted analyses of genes within co-expression modules, we were able to determine functions for these modules that were cell lineage-specific or shared among multiple cell lineages. In addition, our analyses identified transcription factors likely important for immune cell lineage commitment and/or maintenance.
Collapse
Affiliation(s)
- Saraï Mola
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - Sylvain Foisy
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - Gabrielle Boucher
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - François Major
- Unité de recherche en ingénierie des ARN, Institut de recherche en immunologie et en cancérologie, Montréal, Québec, Canada
- Département d’informatique et de recherche opérationnelle, Université de Montréal, Montréal, Québec, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Québec, Canada
| | - Claudine Beauchamp
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - Mohamad Karaky
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - Philippe Goyette
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - Sylvie Lesage
- Centre de recherche, Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Québec, Canada
| | - John D. Rioux
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
11
|
Andersen BJ, Rosa BA, Kupritz J, Meite A, Serge T, Hertz MI, Curtis K, King CL, Mitreva M, Fischer PU, Weil GJ. Systems analysis-based assessment of post-treatment adverse events in lymphatic filariasis. PLoS Negl Trop Dis 2019; 13:e0007697. [PMID: 31557154 PMCID: PMC6762072 DOI: 10.1371/journal.pntd.0007697] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/12/2019] [Indexed: 12/20/2022] Open
Abstract
Background Lymphatic filariasis (LF) is a neglected tropical disease, and the Global Program to Eliminate LF delivers mass drug administration (MDA) to 500 million people every year. Adverse events (AEs) are common after LF treatment. Methodology/Principal findings To better understand the pathogenesis of AEs, we studied LF-patients from a treatment trial. Plasma levels of many filarial antigens increased post-treatment in individuals with AEs, and this is consistent with parasite death. Circulating immune complexes were not elevated in these participants, and the classical complement cascade was not activated. Multiple cytokines increased after treatment in persons with AEs. A transcriptomic analysis was performed for nine individuals with moderate systemic AEs and nine matched controls. Differential gene expression analysis identified a significant transcriptional signature associated with post-treatment AEs; 744 genes were upregulated. The transcriptional signature was enriched for TLR and NF-κB signaling. Increased expression of seven out of the top eight genes upregulated in persons with AEs were validated by qRT-PCR, including TLR2. Conclusions/Significance This is the first global study of changes in gene expression associated with AEs after treatment of lymphatic filariasis. Changes in cytokines were consistent with prior studies and with the RNAseq data. These results suggest that Wolbachia lipoprotein is involved in AE development, because it activates TLR2-TLR6 and downstream NF-κB. Additionally, LPS Binding Protein (LBP, which shuttles lipoproteins to TLR2) increased post-treatment in individuals with AEs. Improved understanding of the pathogenesis of AEs may lead to improved management, increased MDA compliance, and accelerated LF elimination. Lymphatic filariasis (LF) is a disabling parasitic disease that affects millions of people in the developing world. The Global Programme to Eliminate Lymphatic Filariasis (coordinated by the World Health Organization) uses mass administration of antifilarial medications to cure infections, prevent disease, and reduce transmission. Some individuals develop adverse events (AEs) after treatment, and this can reduce willingness of persons in endemic areas to accept treatment. The purpose of this study was to improve understanding of the cause of AEs following treatment. We hypothesized that parasite antigens released into the blood following treatment trigger inflammatory responses that lead to AEs. To test this hypothesis we collected blood from LF-infected individuals before and after treatment and clinically assessed them for AEs. We measured parasite antigens, cytokines and other components of the immune system in blood samples and compared post-treatment changes in persons with and without AEs. We also assessed changes in transcription profiles in peripheral blood leukocytes that were associated with post-treatment AEs. Post-treatment changes in transcription profiles and in immune proteins and parasite components in plasma suggest that systemic AEs are triggered by death of the parasites following treatment with release of parasite antigens and Wolbachia bacteria into the circulation. Improved understanding of the pathogenesis of post-treatment AEs may help to improve messaging related to mass drug administration programs and lead to improved AE management.
Collapse
Affiliation(s)
- Britt J. Andersen
- Infectious Diseases Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Bruce A. Rosa
- McDonnell Genome Institute, Washington University School of Medicine, St.Louis, Missouri, United States of America
| | - Jonah Kupritz
- Infectious Diseases Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Aboulaye Meite
- Programme National de la Lutte Contre la Schistosomiase, Les Geohelminthiases et la Filariose Lymphatique, Abidjan, Côte d’Ivoire
| | - Traye Serge
- Programme National de la Lutte Contre la Schistosomiase, Les Geohelminthiases et la Filariose Lymphatique, Abidjan, Côte d’Ivoire
| | - Marla I. Hertz
- Infectious Diseases Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kurt Curtis
- Infectious Diseases Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Christopher L. King
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Makedonka Mitreva
- Infectious Diseases Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- McDonnell Genome Institute, Washington University School of Medicine, St.Louis, Missouri, United States of America
| | - Peter U. Fischer
- Infectious Diseases Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Gary J. Weil
- Infectious Diseases Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
12
|
Estelius J, Lengqvist J, Ossipova E, Idborg H, Le Maître E, Andersson MLA, Brundin L, Khademi M, Svenungsson E, Jakobsson PJ, Lampa J. Mass spectrometry-based analysis of cerebrospinal fluid from arthritis patients-immune-related candidate proteins affected by TNF blocking treatment. Arthritis Res Ther 2019; 21:60. [PMID: 30770760 PMCID: PMC6377734 DOI: 10.1186/s13075-019-1846-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/06/2019] [Indexed: 12/16/2022] Open
Abstract
Background Signs of inflammation in cerebrospinal fluid (CSF) of rheumatoid arthritis patients correlate positively with fatigue, a central nervous system (CNS)-related symptom that can be partially suppressed by TNF blockade. This suggests a possible role for CNS inflammation in arthritis that may be affected by TNF blockade. We therefore investigated the effects of TNF blockade on the arthritis CSF proteome and how candidate proteins related to clinical measures of disease activity and inflammation. Methods Mass spectrometry-based quantitative proteomic analysis was performed on CSF from seven polyarthritis patients before and during infliximab treatment. Treatment-associated proteins were identified using univariate (Wilcoxon signed rank test) and multivariate (partial least squares discriminant analysis (PLS-DA)) strategies. Relations between selected candidate proteins and clinical measures were investigated using the Spearman correlations. Additionally, selected proteins were cross-referenced to other studies investigating human CSF in a thorough literature search to ensure feasibility of our results. Results Univariate analysis of arthritis CSF proteome revealed a decrease of 35 proteins, predominantly involved in inflammatory processes, following TNF blockade. Seven candidate proteins, Contactin-1 (CNTN1), fibrinogen gamma chain (FGG), hemopexin (HPX), cell adhesion molecule-3 (CADM3), alpha-1B-glycoprotein (A1BG), complement factor B (CFB), and beta-2-microglobulin (B2M), were selected for further studies based on identification by both univariate and multivariate analyses and reported detection in human CSF and known associations to arthritis. Decreased levels of FGG and CFB in CSF after treatment showed strong correlations with both erythrocyte sedimentation rate and disability scores, while CNTN1 and CADM3 were associated with pain. Conclusion Several immune-related proteins in the CSF of arthritis patients decreased during TNF blockade, including FGG and CFB that both correlated strongly with systemic inflammation. Our findings stress that also intrathecal inflammatory pathways are related to arthritis symptoms and may be affected by TNF blockade. Electronic supplementary material The online version of this article (10.1186/s13075-019-1846-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Johanna Estelius
- Rheumatology Unit, Department of Medicine, Solna, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Johan Lengqvist
- Rheumatology Unit, Department of Medicine, Solna, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Elena Ossipova
- Rheumatology Unit, Department of Medicine, Solna, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Helena Idborg
- Rheumatology Unit, Department of Medicine, Solna, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Erwan Le Maître
- Rheumatology Unit, Department of Medicine, Solna, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Magnus L A Andersson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Lou Brundin
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Mohsen Khademi
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Elisabet Svenungsson
- Rheumatology Unit, Department of Medicine, Solna, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Solna, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Jon Lampa
- Rheumatology Unit, Department of Medicine, Solna, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden.
| |
Collapse
|
13
|
Bohn T, Planchon S, Leclercq CC, Renaut J, Mihaly J, Beke G, Rühl R. Proteomic responses of carotenoid and retinol administration to Mongolian gerbils. Food Funct 2018; 9:3835-3844. [PMID: 29951678 DOI: 10.1039/c8fo00278a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Various health benefits of carotenoids have been described. However, while human observational studies generally suggest positive health effects, supplementation with relatively high doses of individual carotenoids (supplements) have partly produced adverse effects. In the present study, we investigated the effect of several carotenoids on the proteomic response of male Mongolian gerbils (aged 6 weeks). Five groups of gerbils (n = 6 per group) received either retinol (vitamin A/53 mg per kg bw), all-trans β-carotene (pro-vitamin A/100 mg kg-1), the non-pro vitamin A carotenoid lutein (100 mg kg-1), the acyclic carotenoid lycopene (100 mg kg-1) or vehicle (Cremophor EL), via oral single gavage. Gerbils were 12 h post-prandially sacrificed and blood plasma, liver, and white adipose tissue were collected. For liver and adipose tissue, a 2D-DIGE (difference gel electrophoresis) approach was conducted; for plasma, proteomic analyses were achieved by liquid chromatography-mass spectrometry. Compared to controls (vehicle), various proteins were showing significant abundance variations in plasma (66), liver (29) and adipose tissue (19), especially regarding structure (22), protein metabolism (15) and immune system/inflammation (19) functions, while proteins related to antioxidant effects were generally less abundant, suggesting no in vivo relevance. Surprisingly, a large overlap in protein regulation was found between lycopene and retinol exposure, while other carotenoids, including all-trans β-carotene, did not show this overlap. Mainly retinoid acid receptor co-regulated proteins may mechanistically explain this overlapping regulation. This overlapping regulation may be related to common nuclear hormone receptor mediated signalling, though further studies using synthetic ligands of retinoid receptors targeting protein regulation are needed for confirmation.
Collapse
Affiliation(s)
- Torsten Bohn
- Luxembourg Institute of Health, Population Health Department, 1 A-B, rue Thomas Edison, L-1445 Strassen, Luxembourg.
| | | | | | | | | | | | | |
Collapse
|
14
|
Su S, Chen J, Yao H, Liu J, Yu S, Lao L, Wang M, Luo M, Xing Y, Chen F, Huang D, Zhao J, Yang L, Liao D, Su F, Li M, Liu Q, Song E. CD10 +GPR77 + Cancer-Associated Fibroblasts Promote Cancer Formation and Chemoresistance by Sustaining Cancer Stemness. Cell 2018; 172:841-856.e16. [PMID: 29395328 DOI: 10.1016/j.cell.2018.01.009] [Citation(s) in RCA: 855] [Impact Index Per Article: 122.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/28/2017] [Accepted: 01/04/2018] [Indexed: 12/12/2022]
Abstract
Carcinoma-associated fibroblasts (CAFs) are abundant and heterogeneous stromal cells in tumor microenvironment that are critically involved in cancer progression. Here, we demonstrate that two cell-surface molecules, CD10 and GPR77, specifically define a CAF subset correlated with chemoresistance and poor survival in multiple cohorts of breast and lung cancer patients. CD10+GPR77+ CAFs promote tumor formation and chemoresistance by providing a survival niche for cancer stem cells (CSCs). Mechanistically, CD10+GPR77+ CAFs are driven by persistent NF-κB activation via p65 phosphorylation and acetylation, which is maintained by complement signaling via GPR77, a C5a receptor. Furthermore, CD10+GPR77+ CAFs promote successful engraftment of patient-derived xenografts (PDXs), and targeting these CAFs with a neutralizing anti-GPR77 antibody abolishes tumor formation and restores tumor chemosensitivity. Our study reveals a functional CAF subset that can be defined and isolated by specific cell-surface markers and suggests that targeting the CD10+GPR77+ CAF subset could be an effective therapeutic strategy against CSC-driven solid tumors.
Collapse
Affiliation(s)
- Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jianing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Herui Yao
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Shubin Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Liyan Lao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Minghui Wang
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Manli Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yue Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Fei Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Di Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jinghua Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Linbin Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Dan Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Fengxi Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Mengfeng Li
- Department of Microbiology and Key Laboratory of Tropical Disease Control , Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Program of Molecular Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
15
|
AICAR suppresses TNF-α-induced complement factor B in RPE cells. Sci Rep 2017; 7:17651. [PMID: 29247196 PMCID: PMC5732305 DOI: 10.1038/s41598-017-17744-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 11/29/2017] [Indexed: 11/08/2022] Open
Abstract
Age related macular degeneration is the leading cause of blindness in the developed world. Although its precise cause remains elusive, dysfunction of the retinal pigment epithelium (RPE) and dysregulation of complement have been implicated in its pathogenesis. The goal of this study was to evaluate the role of an AMP-dependent kinase (AMPK) activator, 5-aminoimidazole-4-carboxamide riboside (AICAR), on tumor necrosis factor alpha (TNF-α) induction of complement factor B (CFB) in RPE cells. We found that AICAR inhibited TNF-α-induced CFB expression in ARPE-19 and human primary RPE cells in a dose-dependent fashion. Treatment of cells with dipyridamole, which blocks AICAR cellular uptake abolished these effects. In contrast, the adenosine kinase inhibitor, 5-iodotubericidin, which inhibits the conversion of AICAR to the direct activator of AMPK, ZMP, did not reverse the effects on TNF-α-induced CFB expression, suggesting AMPK-independent effects. Indeed, knockout of AMPK in RPE cells using Clustered Regularly Interspaced Palindromic Repeats (CRISPR)/Cas9 did not abolish the inhibitory effects of AICAR on RPE CFB expression. Collectively, our results suggest that AICAR can suppress TNF-α-induced CFB expression in RPE cells in an AMPK-independent mechanism, and could be used as a therapeutic target in certain complement over-activation scenarios.
Collapse
|
16
|
Casiraghi F, Azzollini N, Todeschini M, Fiori S, Cavinato RA, Cassis P, Solini S, Pezzuto F, Mister M, Thurman JM, Benigni A, Remuzzi G, Noris M. Complement Alternative Pathway Deficiency in Recipients Protects Kidney Allograft From Ischemia/Reperfusion Injury and Alloreactive T Cell Response. Am J Transplant 2017; 17:2312-2325. [PMID: 28276660 DOI: 10.1111/ajt.14262] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/26/2017] [Accepted: 02/17/2017] [Indexed: 01/25/2023]
Abstract
Despite the introduction of novel and more targeted immunosuppressive drugs, the long-term survival of kidney transplants has not improved satisfactorily. Early antigen-independent intragraft inflammation plays a critical role in the initiation of the alloimmune response and impacts long-term graft function. Complement activation is a key player both in ischemia/reperfusion injury (IRI) as well as in adaptive antigraft immune response after kidney transplantation. Since the alternative pathway (AP) amplifies complement activation regardless of the initiation pathways and renal IR injured cells undergo uncontrolled complement activation, we speculated whether selective blockade of AP could be a strategy for prolonging kidney graft survival. Here we showed that Balb/c kidneys transplanted in factor b deficient C57 mice underwent reduced IRI and diminished T cell-mediated rejection. In in vitro studies, we found that fb deficiency in T cells and dendritic cells conferred intrinsic impaired alloreactive/allostimulatory functions, respectively, both in direct and indirect pathways of alloantigen presentation. By administering anti-fB antibody to C57 wt recipients in the early post Balb/c kidney transplant phases, we documented that inhibition of AP during both ischemia/reperfusion and early adaptive immune response is necessary for prolonging graft survival. These findings may have implication for the use of AP inhibitors in clinical kidney transplantation.
Collapse
Affiliation(s)
- F Casiraghi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Chiara Cucchi de Alessandri e Gilberto Crespi Transplant Research Center, Bergamo, Italy
| | - N Azzollini
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Chiara Cucchi de Alessandri e Gilberto Crespi Transplant Research Center, Bergamo, Italy
| | - M Todeschini
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Chiara Cucchi de Alessandri e Gilberto Crespi Transplant Research Center, Bergamo, Italy
| | - S Fiori
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Chiara Cucchi de Alessandri e Gilberto Crespi Transplant Research Center, Bergamo, Italy
| | - R A Cavinato
- Laboratory of Clinical and Experimental Immunology, Immunology Department, University of São Paulo, São Paulo, Brazil
| | - P Cassis
- IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - S Solini
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Chiara Cucchi de Alessandri e Gilberto Crespi Transplant Research Center, Bergamo, Italy
| | - F Pezzuto
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Chiara Cucchi de Alessandri e Gilberto Crespi Transplant Research Center, Bergamo, Italy
| | - M Mister
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Chiara Cucchi de Alessandri e Gilberto Crespi Transplant Research Center, Bergamo, Italy
| | - J M Thurman
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - A Benigni
- IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - G Remuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Chiara Cucchi de Alessandri e Gilberto Crespi Transplant Research Center, Bergamo, Italy.,IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy.,Unit of Nephrology and Dialysis, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy.,Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - M Noris
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Chiara Cucchi de Alessandri e Gilberto Crespi Transplant Research Center, Bergamo, Italy
| |
Collapse
|
17
|
Sheen JH, Strainic MG, Liu J, Zhang W, Yi Z, Medof ME, Heeger PS. TLR-Induced Murine Dendritic Cell (DC) Activation Requires DC-Intrinsic Complement. THE JOURNAL OF IMMUNOLOGY 2017; 199:278-291. [PMID: 28539427 DOI: 10.4049/jimmunol.1700339] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/04/2017] [Indexed: 01/04/2023]
Abstract
Induction of proinflammatory T cell immunity is augmented by innate dendritic cell (DC) maturation commonly initiated by TLR signaling. We demonstrate that ligation of TLR3, TLR4, and TLR9 induces murine DC production of complement components and local production of the anaphylatoxin C5a. In vitro, ex vivo, and in vivo analyses show that TLR-induced DC maturation, as assessed by surface phenotype, expression profiling by gene array, and functional ability to stimulate T cell responses, requires autocrine C3a receptor and C5a receptor (C3ar1/C5ar1) signaling. Studies using bone marrow chimeric animals and Foxp3-GFP/ERT2-Cre/dTomato fate-mapping mice show that TLR-initiated DC autocrine C3ar1/C5ar1 signaling causes expansion of effector T cells and instability of regulatory T cells and contributes to T cell-dependent transplant rejection. Together, our data position immune cell-derived complement production and autocrine/paracrine C3ar1/C5ar1 signaling as crucial intermediary processes that link TLR stimulation to DC maturation and the subsequent development of effector T cell responses.
Collapse
Affiliation(s)
- Joong-Hyuk Sheen
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Institute of Immunology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| | - Michael G Strainic
- Institute of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Jinbo Liu
- Institute of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Weijia Zhang
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Zhengzi Yi
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - M Edward Medof
- Institute of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Peter S Heeger
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029; .,Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Institute of Immunology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| |
Collapse
|
18
|
Complement Factor B Production in Renal Tubular Cells and Its Role in Sodium Transporter Expression During Polymicrobial Sepsis. Crit Care Med 2016; 44:e289-99. [PMID: 26757165 DOI: 10.1097/ccm.0000000000001566] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Toll-like receptors and complement are two components of the innate immunity. Complement factor B is essential for the alternative pathway of complement activation. We have recently reported that complement factor B is significantly up-regulated in the kidney and may contribute to acute tubular injury in an animal model of sepsis. This study investigates the mechanisms responsible for the complement factor B up-regulation and its role in sodium transporter expression in tubular cells during sepsis. DESIGN Animal study. SETTING Laboratory investigation. SUBJECTS C57BL/6 J wild-type, complement factor B(-/-), and Nfkb1(tm1Bal) p50(-/-) mice. INTERVENTIONS Human proximal tubular cells and mouse tubular epithelial cells were stimulated with Toll-like receptor agonists. Bay 11-7082 was used to block nuclear factor-κB pathway. Alternative pathway activation was detected by C3 zymosan deposition. Polymicrobial sepsis was created by cecal ligation and puncture. Sodium transporter gene expression was determined by quantitative reverse transcriptase-polymerase chain reaction. MEASUREMENTS AND MAIN RESULTS The agonists for Toll-like receptor 4 (lipopolysaccharide) or Toll-like receptor 3 (polyinosinic-polycytidylic acid) induced a marked increase in complement factor B expression in human proximal tubular cells and mouse tubular epithelial cells both at gene and protein levels. The Toll-like receptor 1/2 agonist, Pam3cys, induced complement factor B production only in human proximal tubular cells, not in mouse tubular epithelial cells. The Toll-like receptor 9 ligand, CpG oligodeoxynucleotides failed to induce complement factor B production either in human proximal tubular cells or in mouse tubular epithelial cells. Lipopolysaccharide/polyinosinic-polycytidylic acid-induced complement factor B up-regulation was blocked by Bay 11-7082, a potent inhibitor of nuclear factor-κB signaling, and in mouse tubular epithelial cells deficient in p50 subunit of nuclear factor-κB. Media from the lipopolysaccharide-treated mouse tubular epithelial cell cultures contained de novo synthesized complement factor B and led to functional alternative pathway activation. In a cecal ligation and puncture model, wild-type septic mice had down-regulated expression of sodium transporters in the kidney compared with the sham. In comparison, complement factor B mice or mice treated with anti-complement factor B displayed preserved levels of Na⁺/K⁺ ATPase-α1 following sepsis. CONCLUSIONS 1) Toll-like receptor 3/4 activation is sufficient to induce complement factor B production via nuclear factor-κB pathway and to enhance alternative pathway activation in the kidney tubular epithelial cells. 2) Complement factor B may contribute to the down-regulation of certain sodium transporter expression during sepsis.
Collapse
|
19
|
Shah S, King EM, Mostafa MM, Altonsy MO, Newton R. DUSP1 Maintains IRF1 and Leads to Increased Expression of IRF1-dependent Genes: A MECHANISM PROMOTING GLUCOCORTICOID INSENSITIVITY. J Biol Chem 2016; 291:21802-21816. [PMID: 27551049 DOI: 10.1074/jbc.m116.728964] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 08/19/2016] [Indexed: 11/06/2022] Open
Abstract
Although the mitogen-activated protein kinase (MAPK) phosphatase, DUSP1, mediates dexamethasone-induced repression of MAPKs, 14 of 46 interleukin-1β (IL1B)-induced mRNAs were significantly enhanced by DUSP1 overexpression in pulmonary A549 cells. These include the interferon regulatory factor, IRF1, and the chemokine, CXCL10. Of these, DUSP1-enhanced mRNAs, 10 including CXCL10, were IRF1-dependent. MAPK inhibitors and DUSP1 overexpression prolonged IRF1 expression by elevating transcription and increasing IRF1 mRNA and protein stability. Conversely, DUSP1 silencing increased IL1B-induced MAPK phosphorylation while significantly reducing IRF1 protein expression at 4 h. This confirms a regulatory network whereby DUSP1 switches off MAPKs to maintain IRF1 expression. There was no repression of IRF1 expression by dexamethasone in primary human bronchial epithelial cells, and in A549 cells IL1B-induced IRF1 protein was only modestly and transiently repressed. Although dexamethasone did not repress IL1B-induced IRF1 protein expression at 4-6 h, silencing of IL1B plus dexamethasone-induced DUSP1 significantly reduced IRF1 expression. IL1B-induced expression of CXCL10 was largely insensitive to dexamethasone, whereas other DUSP1-enhanced, IRF1-dependent mRNAs showed various degrees of repression. With IL1B plus dexamethasone, CXCL10 expression was also IRF1-dependent, and expression was reduced by DUSP1 silencing. Thus, IL1B plus dexamethasone-induced DUSP1 maintains expression of IRF1 and the IRF1-dependent gene, CXCL10. This is supported by chromatin immunoprecipitation showing IRF1 recruitment to be essentially unaffected by dexamethasone at the CXCL10 promoter or at the promoters of more highly repressed IRF1-dependent genes. Since IRF1-dependent genes, such as CXCL10, are central to host defense, these data may help explain the reduced effectiveness of glucocorticoids during asthma exacerbations.
Collapse
Affiliation(s)
- Suharsh Shah
- From the Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada T2N 4Z6 and
| | - Elizabeth M King
- From the Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada T2N 4Z6 and
| | - Mahmoud M Mostafa
- From the Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada T2N 4Z6 and
| | - Mohammed O Altonsy
- From the Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada T2N 4Z6 and.,Department of Zoology, Sohag University, Sohag 825224, Egypt
| | - Robert Newton
- From the Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada T2N 4Z6 and
| |
Collapse
|
20
|
Yamanaka K, Kakuta Y, Miyagawa S, Nakazawa S, Kato T, Abe T, Imamura R, Okumi M, Maeda A, Okuyama H, Mizuno M, Nonomura N. Depression of Complement Regulatory Factors in Rat and Human Renal Grafts Is Associated with the Progress of Acute T-Cell Mediated Rejection. PLoS One 2016; 11:e0148881. [PMID: 26928779 PMCID: PMC4771804 DOI: 10.1371/journal.pone.0148881] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/25/2016] [Indexed: 01/02/2023] Open
Abstract
Background The association of complement with the progression of acute T cell mediated rejection (ATCMR) is not well understood. We investigated the production of complement components and the expression of complement regulatory proteins (Cregs) in acute T-cell mediated rejection using rat and human renal allografts. Methods We prepared rat allograft and syngeneic graft models of renal transplantation. The expression of Complement components and Cregs was assessed in the rat grafts using quantitative real-time PCR (qRT-PCR) and immunofluorescent staining. We also administered anti-Crry and anti-CD59 antibodies to the rat allograft model. Further, we assessed the relationship between the expression of membrane cofactor protein (MCP) by immunohistochemical staining in human renal grafts and their clinical course. Results qRT-PCR results showed that the expression of Cregs, CD59 and rodent-specific complement regulator complement receptor 1-related gene/protein-y (Crry), was diminished in the rat allograft model especially on day 5 after transplantation in comparison with the syngeneic model. In contrast, the expression of complement components and receptors: C3, C3a receptor, C5a receptor, Factor B, C9, C1q, was increased, but not the expression of C4 and C5, indicating a possible activation of the alternative pathway. When anti-Crry and anti-CD59 mAbs were administered to the allograft, the survival period for each group was shortened. In the human ATCMR cases, the group with higher MCP expression in the grafts showed improved serum creatinine levels after the ATCMR treatment as well as a better 5-year graft survival rate. Conclusions We conclude that the expression of Cregs in allografts is connected with ATCMR. Our results suggest that controlling complement activation in renal grafts can be a new strategy for the treatment of ATCMR.
Collapse
Affiliation(s)
- Kazuaki Yamanaka
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoichi Kakuta
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- * E-mail: (YK); (SM)
| | - Shuji Miyagawa
- Division of Organ Transplantation, Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- * E-mail: (YK); (SM)
| | - Shigeaki Nakazawa
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Taigo Kato
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Toyofumi Abe
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryoichi Imamura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masayoshi Okumi
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Akira Maeda
- Division of Organ Transplantation, Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroomi Okuyama
- Division of Organ Transplantation, Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masashi Mizuno
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
21
|
Differential proteomic analysis of virus-enriched fractions obtained from plasma pools of patients with dengue fever or severe dengue. BMC Infect Dis 2015; 15:518. [PMID: 26572220 PMCID: PMC4647599 DOI: 10.1186/s12879-015-1271-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/06/2015] [Indexed: 11/10/2022] Open
Abstract
Background Dengue is the most widespread mosquito-borne viral disease of public health concern. In some patients, endothelial cell and platelet dysfunction lead to life-threatening hemorrhagic dengue fever or dengue shock syndrome. Prognostication of disease severity is urgently required to improve patient management. The pathogenesis of severe dengue has not been fully elucidated, and the role of host proteins associated with viral particles has received little exploration. Methods The proteomes of virion-enriched fractions purified from plasma pools of patients with dengue fever or severe dengue were compared. Virions were purified by ultracentrifugation combined with a water-insoluble polyelectrolyte-based technique. Following in-gel hydrolysis, peptides were analyzed by nano-liquid chromatography coupled to ion trap mass spectrometry and identified using data libraries. Results Both dengue fever and severe dengue viral-enriched fractions contained identifiable viral envelope proteins and host cellular proteins. Canonical pathway analysis revealed the identified host proteins are mainly involved in the coagulation cascade, complement pathway or acute phase response signaling pathway. Some host proteins were over- or under-represented in plasma from patients with severe dengue compared to patients with dengue fever. ELISAs were used to validate differential expression of a selection of identified host proteins in individual plasma samples of patients with dengue fever compared to patients with severe dengue. Among 22 host proteins tested, two could differentiate between dengue fever and severe dengue in two independent cohorts (olfactomedin-4: area under the curve (AUC), 0.958; and platelet factor-4: AUC, 0.836). Conclusion A novel technique of virion-enrichment from plasma has allowed to identify two host proteins that have prognostic value for classifying patients with acute dengue who are more likely to develop a severe dengue. The impact of these host proteins on pathogenicity and disease outcome are discussed. Electronic supplementary material The online version of this article (doi:10.1186/s12879-015-1271-7) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Zhao T, Gao J, Van J, To E, Wang A, Cao S, Cui JZ, Guo JP, Lee M, McGeer PL, Matsubara JA. Age-related increases in amyloid beta and membrane attack complex: evidence of inflammasome activation in the rodent eye. J Neuroinflammation 2015; 12:121. [PMID: 26104676 PMCID: PMC4486438 DOI: 10.1186/s12974-015-0337-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/04/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The membrane attack complex (MAC) is a key player in the pathogenesis of age-related macular degeneration (AMD) and is a putative activator of the NLRP3 inflammasome. Amyloid beta (Aβ), a component of drusen deposits, has also been implicated in inflammasome activation by our work and those of others. However, the interactions of MAC and Aβ are still poorly understood, especially their roles in aging and retinal degenerative pathologies. Since inflammasome activation may represent a key cellular pathway underlying age-related chronic inflammation in the eye, the purpose of this study is to identify the effects associated with MAC and inflammasome activation in the retinal pigment epithelium (RPE)/choroid and to evaluate the therapeutic merits of MAC suppression. METHODS Adult Long-Evans rats were divided into treatment and control groups. Treatment groups received oral aurin tricarboxylic acid complex (ATAC), a MAC inhibitor, in drinking-water, and control groups received drinking-water alone (No ATAC). Groups were sacrificed at 7.5 or 11.5 months, after approximately 40 days of ATAC treatment. To study age-related changes of Aβ and MAC in RPE/choroid, naive animals were sacrificed at 2.5, 7.5, and 11.5 months. Eye tissues underwent immunohistochemistry and western blot analysis for MAC, Aβ, NF-κB activation, as well as cleaved caspase-1 and IL-18. Vitreal samples were collected and assessed by multiplex assays for secreted levels of IL-18 and IL-1β. Statistical analyses were performed, and significance level was set at p ≤ 0.05. RESULTS In vivo studies demonstrated an age-dependent increase in MAC, Aβ, and NF-κB activation in the RPE/choroid. Systemic ATAC resulted in a prominent reduction in MAC formation and a concomitant reduction in inflammasome activation measured by cleaved caspase-1 and secreted levels of IL-18 and IL-1β, but not in NF-κB activation. In vitro studies demonstrated Aβ-induced MAC formation on RPE cells. CONCLUSIONS Age-dependent increases in Aβ and MAC are present in the rodent outer retina. Our results suggest that suppressing MAC formation and subsequent inflammasome activation in the RPE/choroid may reduce chronic low-grade inflammation associated with IL-18 and IL-1β in the outer retina.
Collapse
Affiliation(s)
- Tom Zhao
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, 2550 Willow Street, Vancouver, V5Z 3N9, BC, Canada.
| | - Jiangyuan Gao
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, 2550 Willow Street, Vancouver, V5Z 3N9, BC, Canada.
| | - Jenifer Van
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, 2550 Willow Street, Vancouver, V5Z 3N9, BC, Canada.
| | - Eleanor To
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, 2550 Willow Street, Vancouver, V5Z 3N9, BC, Canada.
| | - Aikun Wang
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, 2550 Willow Street, Vancouver, V5Z 3N9, BC, Canada.
| | - Sijia Cao
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, 2550 Willow Street, Vancouver, V5Z 3N9, BC, Canada.
| | - Jing Z Cui
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, 2550 Willow Street, Vancouver, V5Z 3N9, BC, Canada.
| | - Jian-Ping Guo
- Kinsmen Lab of Neurological Research, University of British Columbia, Vancouver, BC, Canada.
| | - Moonhee Lee
- Kinsmen Lab of Neurological Research, University of British Columbia, Vancouver, BC, Canada.
| | - Patrick L McGeer
- Kinsmen Lab of Neurological Research, University of British Columbia, Vancouver, BC, Canada.
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, 2550 Willow Street, Vancouver, V5Z 3N9, BC, Canada.
| |
Collapse
|
23
|
Surace L, Lysenko V, Fontana AO, Cecconi V, Janssen H, Bicvic A, Okoniewski M, Pruschy M, Dummer R, Neefjes J, Knuth A, Gupta A, van den Broek M. Complement is a central mediator of radiotherapy-induced tumor-specific immunity and clinical response. Immunity 2015; 42:767-77. [PMID: 25888260 DOI: 10.1016/j.immuni.2015.03.009] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 01/16/2015] [Accepted: 03/21/2015] [Indexed: 01/21/2023]
Abstract
Radiotherapy induces DNA damage and cell death, but recent data suggest that concomitant immune stimulation is an integral part of the therapeutic action of ionizing radiation. It is poorly understood how radiotherapy supports tumor-specific immunity. Here we report that radiotherapy induced tumor cell death and transiently activated complement both in murine and human tumors. The local production of pro-inflammatory anaphylatoxins C3a and C5a was crucial to the tumor response to radiotherapy and concomitant stimulation of tumor-specific immunity. Dexamethasone, a drug frequently given during radiotherapy, limited complement activation and the anti-tumor effects of the immune system. Overall, our findings indicate that anaphylatoxins are key players in radiotherapy-induced tumor-specific immunity and the ensuing clinical responses.
Collapse
Affiliation(s)
- Laura Surace
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Veronika Lysenko
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Andrea Orlando Fontana
- Department of Radio-Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Hans Janssen
- Division Cell Biology II, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Antonela Bicvic
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Michal Okoniewski
- ID Scientific IT Services, Swiss Federal Institute for Technology (ETH), Weinbergstrasse 11, 8092 Zurich, Switzerland
| | - Martin Pruschy
- Department of Radio-Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Jacques Neefjes
- Division Cell Biology II, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Alexander Knuth
- Clinic of Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Anurag Gupta
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
24
|
Lee MJ, Na K, Jeong SK, Lim JS, Kim SA, Lee MJ, Song SY, Kim H, Hancock WS, Paik YK. Identification of human complement factor B as a novel biomarker candidate for pancreatic ductal adenocarcinoma. J Proteome Res 2014; 13:4878-88. [PMID: 25057901 DOI: 10.1021/pr5002719] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pancreatic cancer (PC; pancreatic ductal adenocarcinoma) is characterized by significant morbidity and mortality worldwide. Although carbohydrate antigen (CA) 19-9 has been known as a PC biomarker, it is not commonly used for general screening because of its low sensitivity and specificity. Therefore, there is an urgent need to develop a new biomarker for PC diagnosis in the earlier stage of cancer. To search for a novel serologic PC biomarker, we carried out an integrated proteomic analysis for a total of 185 pooled or individual plasma from healthy donors and patients with five disease groups including chronic pancreatitis (CP), PC, and other cancers (e.g., hepatocellular carcinoma, cholangiocarcinoma, and gastric cancer) and identified complement factor b (CFB) as a candidate serologic biomarker for PC diagnosis. Immunoblot analysis of CFB revealed more than two times higher expression in plasma samples from PC patients compared with plasma from individuals without PC. Immunoprecipitation coupled to mass spectrometry analysis confirmed both molecular identity and higher expression of CFB in PC samples. CFB showed distinctly higher specificity than CA 19-9 for PC against other types of digestive cancers and in discriminating PC patients from non-PC patients (p < 0.0001). In receiver operator characteristic curve analysis, CFB showed an area under curve of 0.958 (95% CI: 0.956 to 0.959) compared with 0.833 (95% CI: 0.829 to 0.837) for CA 19-9. Furthermore, the Y-index of CFB was much higher than that of CA 19-9 (71.0 vs 50.4), suggesting that CFB outperforms CA 19-9 in discriminating PC from CP and other gastrointestinal cancers. This was further supported by immunoprecipitation and qRT-PCR assays showing higher expression of CFB in PC cell lines than in normal cell lines. A combination of CFB and CA 19-9 showed markedly improved sensitivity (90.1 vs 73.1%) over that of CFB alone in the diagnosis of PC against non-PC, with similar specificity (97.2 vs 97.9%). Thus, our results identify CFB as a novel serologic PC biomarker candidate and warrant further investigation into a large-scale validation and its role in molecular mechanism of pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Min Jung Lee
- Yonsei Proteome Research Center and ‡Department of Integrated OMICS for Biomedical Science and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University , 50 Yonsei-ro, Sudaemoon-ku, Seoul 120-749, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Nagi-Miura N, Okuzaki D, Torigata K, Sakurai MA, Ito A, Ohno N, Nojima H. CAWS administration increases the expression of interferon γ and complement factors that lead to severe vasculitis in DBA/2 mice. BMC Immunol 2013; 14:44. [PMID: 24063402 PMCID: PMC3876726 DOI: 10.1186/1471-2172-14-44] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/10/2013] [Indexed: 11/17/2022] Open
Abstract
Background Candida albicans water-soluble fraction (CAWS), a mannoprotein-β-glucan complex obtained from the culture supernatant of C. albicans NBRC1385, causes CAWS-mediated vasculitis (CAWS-vasculitis) in B6 and DBA/2 mice with mild and lethal symptoms, respectively. Why CAWS is lethal only in DBA/2 mice remains unknown. Results We performed DNA microarray analyses using mRNA obtained from peripheral blood mononuclear cells (PBMCs) of B6 and DBA/2 mice and compared their respective transcriptomes. We found that the mRNA levels of interferon-γ (Ifng) and several genes that regulate the complement system, such as C3, C4, Cfb, Cfh, and Fcna, were increased dramatically only in DBA/2 mice at 4 and 8 weeks after CAWS administration. The dramatic increase was confirmed by quantitative real-time polymerase chain reactions (qRT-PCR). Moreover, mRNA levels of immune-related genes, such as Irf1, Irf7, Irf9, Cebpb, Ccl4, Itgam, Icam1, and IL-12rb1, whose expression levels are known to be increased by Ifng, were also increased, but only in DBA/2 mice. By contrast, the mRNA level of Dectin-2, the critical receptor for the α-mannans of CAWS, was increased slightly and similarly in both B6 and DBA/2 mice after CAWS administration. Conclusions Taken together, our results suggest that CAWS administration induces Dectin-2 mediated CAWS-vasculitis in both B6 and DBA/2 mice and the expression of Ifng, but only in DBA/2 mice, which led to increased expression of C3, C4, Cfb, Cfh, and Fcna and an associated increase in lethality in these mice. This model may contribute to our understanding of the pathogenesis of severe human vasculitis.
Collapse
Affiliation(s)
- Noriko Nagi-Miura
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0329, Japan.
| | | | | | | | | | | | | |
Collapse
|
26
|
Luo C, Chen M, Madden A, Xu H. Expression of complement components and regulators by different subtypes of bone marrow-derived macrophages. Inflammation 2013; 35:1448-61. [PMID: 22450524 DOI: 10.1007/s10753-012-9458-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Under inflammatory conditions, macrophages can differentiate into different functional subtypes. We show that bone marrow-derived macrophages constitutively express different levels of various complement-related genes. The relative expression levels are C1qb > Crry > CFH > C3 > C1r > CFB > DAF1 > CD59a > C2 > C1INH > C1s > C4. Upon activation, the expression of C1r, C1s, C3, C2, CFB, and C1INH was up-regulated, and CFH, CD59a, and DAF1, down-regulated in M1 (induced by interferon-γ + lipopolysaccharides (LPS)) and M2b (induced by immune complex + LPS) macrophages. The expression of C4 and CFH was slightly up-regulated in interleukin (IL)-10-induced M2c macrophages. Complement gene expression in IL-4-induced M2a macrophages was weakly down-regulated as compared to resting M0 macrophages. Higher levels of C3, C1INH, and CFB but lower levels of CFH expression in M1 and M2b macrophage suggests that they may be involved in the alternative pathway of complement activation during inflammation.
Collapse
Affiliation(s)
- Chang Luo
- Centre for Vision and Vascular Science, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Grosvenor Road, BT12 6BA, Belfast, UK
| | | | | | | |
Collapse
|
27
|
Kaczorowski DJ, Scott MJ, Pibris JP, Afrazi A, Nakao A, Edmonds RD, Kim S, Kwak JH, Liu Y, Fan J, Billiar TR. Mammalian DNA is an endogenous danger signal that stimulates local synthesis and release of complement factor B. Mol Med 2012; 18:851-60. [PMID: 22526919 DOI: 10.2119/molmed.2012.00011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 04/19/2012] [Indexed: 11/06/2022] Open
Abstract
Complement factor B plays a critical role in ischemic tissue injury and autoimmunity. Factor B is dynamically synthesized and released by cells outside of the liver, but the molecules that trigger local factor B synthesis and release during endogenous tissue injury have not been identified. We determined that factor B is upregulated early after cold ischemia-reperfusion in mice, using a heterotopic heart transplant model. These data suggested upregulation of factor B by damage-associated molecular patterns (DAMPs), but multiple common DAMPs did not induce factor B in RAW264.7 mouse macrophages. However, exogenous DNA induced factor B mRNA and protein expression in RAW cells in vitro, as well as in peritoneal and alveolar macrophages in vivo. To determine the cellular mechanisms involved in DNA-induced factor B upregulation we then investigated the role of multiple known DNA receptors or binding partners. We stimulated peritoneal macrophages from wild-type (WT), toll-like receptor 9 (TLR9)-deficient, receptor for advanced glycation end products (RAGE)⁻/⁻ and myeloid differentiation factor 88 (MyD88)⁻/⁻ mice, or mouse macrophages deficient in high-mobility group box proteins (HMGBs), DNA-dependent activator of interferon-regulatory factors (DAI) or absent in melanoma 2 (AIM2), with DNA in the presence or absence of lipofection reagent. Reverse transcription-polymerase chain reaction, Western blotting and immunocytochemical analysis were employed for analysis. Synthesis of factor B was independent of TLR9, RAGE, DAI and AIM2, but was dependent on HMGBs, MyD88, p38 and NF-κB. Our data therefore show that mammalian DNA is an endogenous molecule that stimulates factor B synthesis and release from macrophages via HMGBs, MyD88, p38 and NF-κB signaling. This activation of the immune system likely contributes to damage following sterile injury such as hemorrhagic shock and ischemia-reperfusion.
Collapse
Affiliation(s)
- David J Kaczorowski
- Division of Cardiovascular Surgery, University of Pennsylvania, Philadelphia, PA, United States of America
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gu W, Chen J, Yang L, Zhao KN. TNF-α promotes IFN-γ-induced CD40 expression and antigen process in Myb-transformed hematological cells. ScientificWorldJournal 2012; 2012:621969. [PMID: 22547990 PMCID: PMC3322478 DOI: 10.1100/2012/621969] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 10/03/2011] [Indexed: 01/10/2023] Open
Abstract
Tumour necrosis factor-α, interferon-γ and interleukin-4 are critical cytokines in regulating the immune responses against infections and tumours. In this study, we investigated the effects of three cytokines on CD40 expression in Myb-transformed hematological cells and their regulatory roles in promoting these cells into dendritic cells. We observed that both interleukin-4 and interferon-γ increased CD40 expression in these hematological cells in a dose-dependent manner, although the concentration required for interleukin-4 was significantly higher than that for interferon-γ. We found that tumour necrosis factor-α promoted CD40 expression induced by interferon-γ, but not by interleukin-4. Our data showed that tumour necrosis factor-α plus interferon-γ-treated Myb-transformed hematological cells had the greatest ability to take up and process the model antigen DQ-Ovalbumin. Tumour necrosis factor-α also increased the ability of interferon-γ to produce the mixed lymphocyte reaction to allogenic T cells. Furthermore, only cotreatment with tumour necrosis factor-α and interferon-γ induced Myb-transformed hematological cells to express interleukin-6. These results suggest that tumour necrosis factor-α plays a key regulatory role in the development of dendritic cells from hematological progenitor cells induced by interferon-γ.
Collapse
Affiliation(s)
- Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | |
Collapse
|
29
|
Zipplies JK, Kirschfink M, Amann B, Hauck SM, Stangassinger M, Deeg CA. Complement factor B expression profile in a spontaneous uveitis model. Immunobiology 2010; 215:949-55. [DOI: 10.1016/j.imbio.2010.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 02/10/2010] [Accepted: 02/10/2010] [Indexed: 10/19/2022]
|
30
|
Segawa Y, Hisano S, Matsushita M, Fujita T, Hirose S, Takeshita M, Iwasaki H. IgG subclasses and complement pathway in segmental and global membranous nephropathy. Pediatr Nephrol 2010; 25:1091-9. [PMID: 20151159 DOI: 10.1007/s00467-009-1439-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2009] [Revised: 12/28/2009] [Accepted: 12/30/2009] [Indexed: 11/28/2022]
Abstract
The aim of our study was to clarify the association between immunoglobulin G(IgG) subclasses and the complement pathway in patients with idiopathic membranous nephropathy (MN). Immunofluorescence (IF) was performed in 16 MN patients and 20 controls using antibodies against IgG, IgA, IgM, C1q, C3c, C4d, IgG1, IgG2, IgG3, IgG4, mannose binding lectin (MBL), C4-binding protein (C4-bp), factor B, C5b-9, and CD59. MN was classified into two types, segmental MN (S-MN; six patients) and global MN (G-MN; ten patients), according to the distribution of IgG deposits along the glomerular capillary wall. No deposition of any antibody was found in the controls. IF revealed IgG1, IgG3, C1q, C3c, C4d, C4-bp, C5b-9, and CD59 deposits in patients with S-MN, whereas IgG1, IgG2, IgG3, IgG4, C3c, C4d, MBL, factor B, C4-bp, C5b-9, and CD59 deposits were detected in those with G-MN. There was a higher deposition of IG1, IgG2, and IgG4 in patients with G-MN than in those with S-MN, whereas the intensity of C1q deposits was higher in S-MN than in G-MN patients. In contrast, the intensity of factor B and MBL was higher in G-MN than in S-MN patients. This is the first report of S-MN patients showing complement activation of the classical pathway associated with IgG1 and IgG3 and G-MN patients showing complement activation of both the alternative and lectin pathways associated with IgG2 and IgG4.
Collapse
Affiliation(s)
- Yoshie Segawa
- Department of Pathology, Faculty of Medicine, Fukuoka University, Nanakuma 7-45-1, Jonan-ku, Fukuoka City, 814-0180, Japan
| | | | | | | | | | | | | |
Collapse
|
31
|
Kaczorowski DJ, Afrazi A, Scott MJ, Kwak JH, Gill R, Edmonds RD, Liu Y, Fan J, Billiar TR. Pivotal advance: The pattern recognition receptor ligands lipopolysaccharide and polyinosine-polycytidylic acid stimulate factor B synthesis by the macrophage through distinct but overlapping mechanisms. J Leukoc Biol 2010; 88:609-18. [PMID: 20413727 DOI: 10.1189/jlb.0809588] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
TLRs and complement are critical to the host response in sepsis, trauma, and ischemia/reperfusion. We hypothesize that TLR stimulation leads to synthesis and release of complement components by macrophages, an important source of extrahepatic complement. RAW264.7 macrophages or peritoneal macrophages from WT and TLR4-, TLR3-, TRIF-, or MyD88-deficient mice were cultured under standard conditions. In some experiments, cells were pretreated with inhibitors of MAPKs or a NF-κB inhibitor. Cells were stimulated with TLR ligands at known stimulatory concentrations. Intratracheal and i.p. injections were also performed in mice. RT-PCR, Western blotting, and immunocytochemistry were used for analysis. Using a RT-PCR-based panel, we demonstrate that of 18 complement components tested, factor B of the alternative pathway is the most robustly up-regulated complement component in macrophages in response to LPS. This up-regulation results in release of factor B into the media. Up-regulation of factor B by LPS is dependent on TLR4, TRIF, JNK, and NF-κB. A screen of other TLR ligands demonstrated that stimulation with poly I:C (dsRNA analog) also results in up-regulation of factor B, which is dependent on JNK and NF-κB but independent of TLR3 and TRIF. Up-regulation of factor B is also observed after intratracheal and i.p. injection of LPS or poly I:C in vivo. PRR stimulation profoundly influences production and release of factor B by macrophages. Understanding the mechanisms of PRR-mediated complement production may lead to strategies aimed at preventing tissue damage in diverse settings, including sepsis, trauma, and ischemia/reperfusion.
Collapse
Affiliation(s)
- David J Kaczorowski
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Acetylation of p65 at lysine 314 is important for late NF-kappaB-dependent gene expression. BMC Genomics 2010; 11:22. [PMID: 20064247 PMCID: PMC2823688 DOI: 10.1186/1471-2164-11-22] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Accepted: 01/11/2010] [Indexed: 02/01/2023] Open
Abstract
Background NF-κB regulates the expression of a large number of target genes involved in the immune and inflammatory response, apoptosis, cell proliferation, differentiation and survival. We have earlier reported that p65, a subunit of NF-κB, is acetylated in vitro and in vivo at three different lysines (K310, K314 and K315) by the histone acetyltransferase p300. Results In this study, we describe that site-specific mutation of p65 at lysines 314 and 315 enhances gene expression of a subset of NF-κB target genes including Mmp10 and Mmp13. Increased gene expression was mainly observed three hours after TNFα stimulation. Chromatin immunoprecipitation (ChIP) experiments with an antibody raised against acetylated lysine 314 revealed that chromatin-bound p65 is indeed acetylated at lysine 314. Conclusions Together, our results establish acetylation of K314 as an important regulatory modification of p65 and subsequently of NF-κB-dependent gene expression.
Collapse
|
33
|
Smith LC, Ghosh J, Buckley KM, Clow LA, Dheilly NM, Haug T, Henson JH, Li C, Lun CM, Majeske AJ, Matranga V, Nair SV, Rast JP, Raftos DA, Roth M, Sacchi S, Schrankel CS, Stensvåg K. Echinoderm Immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 708:260-301. [DOI: 10.1007/978-1-4419-8059-5_14] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
34
|
Wang J, Ohno-Matsui K, Yoshida T, Shimada N, Ichinose S, Sato T, Mochizuki M, Morita I. Amyloid-β up-regulates complement factor B in retinal pigment epithelial cells through cytokines released from recruited macrophages/microglia: Another mechanism of complement activation in age-related macular degeneration. J Cell Physiol 2009; 220:119-28. [DOI: 10.1002/jcp.21742] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
35
|
Nikolskaya T, Nikolsky Y, Serebryiskaya T, Zvereva S, Sviridov E, Dezso Z, Rahkmatulin E, Brennan RJ, Yankovsky N, Bhattacharya SK, Agapova O, Hernandez MR, Shestopalov VI. Network analysis of human glaucomatous optic nerve head astrocytes. BMC Med Genomics 2009; 2:24. [PMID: 19426536 PMCID: PMC2705386 DOI: 10.1186/1755-8794-2-24] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Accepted: 05/09/2009] [Indexed: 12/01/2022] Open
Abstract
Background Astrocyte activation is a characteristic response to injury in the central nervous system, and can be either neurotoxic or neuroprotective, while the regulation of both roles remains elusive. Methods To decipher the regulatory elements controlling astrocyte-mediated neurotoxicity in glaucoma, we conducted a systems-level functional analysis of gene expression, proteomic and genetic data associated with reactive optic nerve head astrocytes (ONHAs). Results Our reconstruction of the molecular interactions affected by glaucoma revealed multi-domain biological networks controlling activation of ONHAs at the level of intercellular stimuli, intracellular signaling and core effectors. The analysis revealed that synergistic action of the transcription factors AP-1, vitamin D receptor and Nuclear Factor-kappaB in cross-activation of multiple pathways, including inflammatory cytokines, complement, clusterin, ephrins, and multiple metabolic pathways. We found that the products of over two thirds of genes linked to glaucoma by genetic analysis can be functionally interconnected into one epistatic network via experimentally-validated interactions. Finally, we built and analyzed an integrative disease pathology network from a combined set of genes revealed in genetic studies, genes differentially expressed in glaucoma and closely connected genes/proteins in the interactome. Conclusion Our results suggest several key biological network modules that are involved in regulating neurotoxicity of reactive astrocytes in glaucoma, and comprise potential targets for cell-based therapy.
Collapse
Affiliation(s)
- Tatiana Nikolskaya
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 3 Gubkina Str, Moscow, Russia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Goring K, Huang Y, Mowat C, Léger C, Lim TH, Zaheer R, Mok D, Tibbles LA, Zygun D, Winston BW. Mechanisms of human complement factor B induction in sepsis and inhibition by activated protein C. Am J Physiol Cell Physiol 2009; 296:C1140-50. [DOI: 10.1152/ajpcell.00071.2009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To investigate the potential role of the local expression of alternative complement factor B (hBf) in human sepsis, we examined the induction of Bf gene expression in human peripheral blood monocytes (PBMCs) from patients with septic shock and the mechanisms of hBf gene regulation by tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and lipopolysaccharide (LPS) in human monocytes. PBMCs from septic shock patients showed increased hBf mRNA expression when compared with control patients. Costimulation with TNF-α and IFN-γ or stimulation with LPS demonstrated a time- and dose-dependent induction of hBf mRNA expression in human PBMCs. A region of the hBf promoter between −735 and +128 bp was found to mediate IFN-γ, TNF-α, and LPS responsiveness as well as the synergistic effect of IFN-γ/TNF-α on hBf promoter activity. Site-directed mutagenesis of a IFN-γ-activation site (GAS) cis element (−90 to −82 bp) abrogated IFN-γ responsiveness. Mutagenesis of a nuclear factor (NF)-κB cis element at −466 to −456 bp abrogated TNF-α and LPS responsiveness of the Bf promoter. Thus hBf gene expression is induced in PBMCs from septic shock patients, and the induction of hBf by IFN-γ, TNF-α, and LPS is through GAS and NF-κB cis-binding sites on the hBf promoter. Furthermore, activated protein C (APC) inhibited LPS-stimulated hBf promoter activity and protein expression in human monocytes suggesting that the beneficial effect of APC therapy in sepsis may in part be due to inhibition of complement induction and/or activation via the alternative pathway.
Collapse
|
37
|
Singh MV, Kapoun A, Higgins L, Kutschke W, Thurman JM, Zhang R, Singh M, Yang J, Guan X, Lowe JS, Weiss RM, Zimmermann K, Yull FE, Blackwell TS, Mohler PJ, Anderson ME. Ca2+/calmodulin-dependent kinase II triggers cell membrane injury by inducing complement factor B gene expression in the mouse heart. J Clin Invest 2009; 119:986-96. [PMID: 19273909 DOI: 10.1172/jci35814] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Accepted: 01/21/2009] [Indexed: 01/04/2023] Open
Abstract
Myocardial Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibition improves cardiac function following myocardial infarction (MI), but the CaMKII-dependent pathways that participate in myocardial stress responses are incompletely understood. To address this issue, we sought to determine the transcriptional consequences of myocardial CaMKII inhibition after MI. We performed gene expression profiling in mouse hearts with cardiomyocyte-delimited transgenic expression of either a CaMKII inhibitory peptide (AC3-I) or a scrambled control peptide (AC3-C) following MI. Of the 8,600 mRNAs examined, 156 were substantially modulated by MI, and nearly half of these showed markedly altered responses to MI with CaMKII inhibition. CaMKII inhibition substantially reduced the MI-triggered upregulation of a constellation of proinflammatory genes. We studied 1 of these proinflammatory genes, complement factor B (Cfb), in detail, because complement proteins secreted by cells other than cardiomyocytes can induce sarcolemmal injury during MI. CFB protein expression in cardiomyocytes was triggered by CaMKII activation of the NF-kappaB pathway during both MI and exposure to bacterial endotoxin. CaMKII inhibition suppressed NF-kappaB activity in vitro and in vivo and reduced Cfb expression and sarcolemmal injury. The Cfb-/- mice were partially protected from the adverse consequences of MI. Our findings demonstrate what we believe is a novel target for CaMKII in myocardial injury and suggest that CaMKII is broadly important for the genetic effects of MI in cardiomyocytes.
Collapse
Affiliation(s)
- Madhu V Singh
- Division of Cardiovascular Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Hyland KA, Brennan R, Olmsted SB, Rojas E, Murphy E, Wang B, Cleary PP. The early interferon response of nasal-associated lymphoid tissue to Streptococcus pyogenes infection. ACTA ACUST UNITED AC 2009; 55:422-31. [PMID: 19243434 DOI: 10.1111/j.1574-695x.2009.00540.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Streptococcus pyogenes is a major causative agent of tonsillitis or pharyngitis in children. Streptococcus pyogenes can persist in tonsils, and one-third of children treated with antibiotics continue to shed streptococci and have recurrent infections. Mouse nasal-associated lymphoid tissue (NALT) is functionally analogous to human oropharyngeal lymphoid tissues, and serves as a model for characterization of the mucosal innate immune response to S. pyogenes. Wild-type S. pyogenes induces transcription of both type I and interferon-gamma (IFN-gamma)-responsive genes, proinflammatory genes and acute-phase response proteins 24 h after intranasal infection. Invasion of NALT and the induction of the interferon response were not dependent on expression of antiphagocytic M protein. Intranasal infection induces a substantial influx of neutrophils into NALT at 24 h, which declines by 48 h after infection. Infection of IFN-gamma(-/-) [IFN-gamma knock-out mouse (GKO)] C57BL/6 mice with wild-type S. pyogenes resulted in local dissemination of bacteria to draining lymph nodes (LN), but did not lead to systemic infection by 48 h after infection. Infected GKO mice had an increased influx of neutrophils into NALT compared with immunocompetent mice. Thus, IFN-gamma-induced responses are required to prevent local dissemination of streptococci to the draining LN.
Collapse
Affiliation(s)
- Kendra A Hyland
- Department of Microbiology, University of Minnesota, Minneapolis, MN 55413, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Tsilimbaris MK, Panagiotoglou TD, Charisis SK, Anastasakis A, Krikonis TS, Christodoulakis E. The use of intravitreal etanercept in diabetic macular oedema. Semin Ophthalmol 2007; 22:75-9. [PMID: 17564925 DOI: 10.1080/08820530701418243] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The aim of this pilot study was to investigate the effect of intravitreal administration of etanercept in refractory diabetic macular edema. Seven patients diagnosed with diabetic macular edema, refractory to previous treatment, were enrolled. They all received 2 consecutive intravitreal injections of 2.5 mg (0, 1 ml) of Etanercept (Enbrel), with a two-week interval. In all patients visual acuity assessment, fundoscopy and fluorescein angiography were performed prior to the first injection, weekly for the first month, as well as 2 and 3 months following the first injection. No adverse reactions or adverse events were noticed in any patient. Analysis of the data indicates a trend for improvement of visual acuity, a slight worsening of hard exudates and fluorescein leakage, while hemorrhages remained stable, 3 months after initiation of therapy. However, no statistical significance has been reached. This small pilot study did not reveal any improvement in the clinical course of patients with refractory diabetic macular edema after the intravitreal injection of etanercept. Further research is warranted in order to obtain conclusive results concerning the role of anti-TNF therapy in diabetic macular edema.
Collapse
Affiliation(s)
- Miltiadis K Tsilimbaris
- Retina Service, Department of Ophthalmology, University Hospital of Heraklion, Crete, Greece.
| | | | | | | | | | | |
Collapse
|
40
|
Hisano S, Matsushita M, Fujita T, Takeshita M, Iwasaki H. Activation of the lectin complement pathway in post-streptococcal acute glomerulonephritis. Pathol Int 2007; 57:351-7. [PMID: 17539966 DOI: 10.1111/j.1440-1827.2007.02107.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The aim of the present study was to elucidate the correlation between complement pathways and clinicopathological findings in post-streptococcal acute glomerulonephritis (PSAGN). Immunohistological staining was performed on renal specimens obtained from 18 patients with PSAGN and 20 controls, using antibodies against IgG, IgA, IgM, C1q, C3c, C4, fibrinogen, factor B, C4-binding protein (C4-bp), C5b-9, CD59, mannose-binding lectin (MBL) and MBL-associated serine protease-1 (MASP-1). Controls showed no deposition of any antibody. In seven patients, glomerular deposits of C3c, C4, factor B, C4-bp, C5b-9, CD59, MBL and MASP-1 were found. In the remaining 11 patients, glomerular deposits of neither C4 nor MBL/MASP-1 were found, and glomerular deposits of C3c, factor B, C5b-9 and CD59 were evident. C4-bp was detected in seven of these 11 patients. Glomerular deposits of fibrinogen were detected in five of seven patients with MBL/MASP-1 deposits and in only two of 11 patients without MBL/MASP-1 deposits. Hematuria was prolonged in three of seven patients with MBL/MASP-1 deposits through follow up, whereas urinalysis was normal in all patients without MBL/MASP-1 deposits. However, the histological indicators were not different between the two groups. To the authors' knowledge this is the first report to show that complement activation through both the alternative and lectin pathways is evident in some patients with PSAGN. Complement activation is promoted in situ in the glomerulus.
Collapse
Affiliation(s)
- Satoshi Hisano
- Department of Pathology, Faculty of Medicine, Fukuoka University, Nanakuma, Fukuoka, Japan.
| | | | | | | | | |
Collapse
|
41
|
Bykov I, Junnikkala S, Pekna M, Lindros KO, Meri S. Effect of chronic ethanol consumption on the expression of complement components and acute-phase proteins in liver. Clin Immunol 2007; 124:213-20. [PMID: 17586095 DOI: 10.1016/j.clim.2007.05.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 04/27/2007] [Accepted: 05/14/2007] [Indexed: 01/10/2023]
Abstract
The complement system can provoke but also participate in the repair of liver injury. Here we investigated by microarray analysis the effect of chronic ethanol consumption on hepatic mRNA expression of complement components and acute-phase proteins in complement C3-deficient (C3(-/-)) and wild-type (C3(+/+)) mice. Up-regulation by ethanol of factor B, C1qA-chain and clusterin but down-regulation of factor H, Masp-2, factor D and the terminal components C6, C8alpha and C9 was seen in both strains. Ethanol up-regulated C2 and down-regulated C4bp only in C3(+/+) mice, while in C3(-/-) mice up-regulation of C1qB-chain and vitronectin was observed. The expression of factor B, C6, C1qB and factor I was lower but that of factor D higher in C3(-/-) than in C3(+/+) mice. Ethanol induced mRNA synthesis of many acute-phase proteins including SPARC and lipocalin-2, but reduced the expression of SAP. The induction of early classical and alternative pathway components and suppression of terminal pathway components and soluble regulators may thus contribute to alcohol-induced liver injury. Lipocalin-2 and SPARC emerge as new candidate markers for early detection of liver damage.
Collapse
Affiliation(s)
- Igor Bykov
- National Public Health Institute, Departments of Mental Health and Alcohol Research, POB 33, 00250 Helsinki, Finland
| | | | | | | | | |
Collapse
|
42
|
Lindenmeyer MT, Kern C, Sparna T, Donauer J, Wilpert J, Schwager J, Porath D, Kreutz C, Timmer J, Merfort I. Microarray analysis reveals influence of the sesquiterpene lactone parthenolide on gene transcription profiles in human epithelial cells. Life Sci 2007; 80:1608-18. [PMID: 17343877 DOI: 10.1016/j.lfs.2007.01.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2006] [Revised: 01/16/2007] [Accepted: 01/20/2007] [Indexed: 11/25/2022]
Abstract
Sesquiterpene lactones are known for their anti-inflammatory activity which has been proven in various assays on DNA, mRNA and protein level. Here we report on the change in the gene expression profile in TNF-alpha stimulated human 293 cells after treatment with parthenolide using a cDNA microarray analysis. Twenty-one of 7028 genes were found to be up- and 18 down-regulated. They encode for chemoattractants, immune system proteins, glycoproteins, metabolism, serine proteinases, and transcription factors. Confirmatory analyses were carried out using quantitative real-time RT-PCR (TaqMan). Additional studies with selected genes revealed the concentration-dependent influence of parthenolide on the expression of these genes.
Collapse
Affiliation(s)
- M T Lindenmeyer
- Institute for Pharmaceutical Sciences, Department of Pharmaceutical Biology and Biotechnology, Albert-Ludwigs-University, Freiburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bolger MS, Ross DS, Jiang H, Frank MM, Ghio AJ, Schwartz DA, Wright JR. Complement levels and activity in the normal and LPS-injured lung. Am J Physiol Lung Cell Mol Physiol 2006; 292:L748-59. [PMID: 17071722 DOI: 10.1152/ajplung.00127.2006] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Complement, a complex protein system, plays an essential role in host defense through bacterial lysis, stimulation of phagocytosis, recruitment of immune cells to infected tissue, and promotion of the inflammatory response. Although complement is most well-characterized in serum, complement activity is also present in the lung. Here we further characterize the complement system in the normal and inflamed lung. By Western blot, C5, C6, and factor I were detected in bronchoalveolar lavage (BAL) at lower levels than in serum, whereas C2 was detected at similar levels in BAL and serum. C4 binding protein (C4BP) was not detectable in BAL. Exposure to lipopolysaccharide (LPS) elevated levels of C1q, factor B, C2, C4, C5, C6, and C3 in human BAL and C3, C5, and factor B in mouse and rat BAL. Message for C1q-B, C1r, C1s, C2, C4, C3, C5, C6, factor B, and factor H, but not C9 or C4BP, was readily detectable by RT-PCR in normal mouse lung. Exposure to LPS enhanced factor B expression, decreased C5 expression, and did not affect C1q-B expression in mouse and rat lung. BAL from rats exposed to LPS had a greater ability to deposit C3b onto bacteria through complement activation than did BAL from control rats. In summary, these data demonstrate that complement levels, expression, and function are altered in acute lung injury and suggest that complement within the lung is regulated to promote opsonization of pathogens and limit potentially harmful inflammation.
Collapse
Affiliation(s)
- Molly S Bolger
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Goldberg M, Luknar-Gabor N, Keidar R, Katz Y. Synthesis of complement proteins in the human chorion is differentially regulated by cytokines. Mol Immunol 2006; 44:1737-42. [PMID: 17005253 DOI: 10.1016/j.molimm.2006.07.298] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 07/13/2006] [Accepted: 07/24/2006] [Indexed: 11/26/2022]
Abstract
The aim of the current paper was to determine the chorion's contribution to complement synthesis in the placenta and its regulation by cytokines. Biosynthetic labeling followed by immunoprecipitation with polyclonal antibodies was performed in chorionic tissue and chorion-derived cells. Eight complement proteins, factor B, C3, C1r, C1s, C1 inhibitor, factor H, C4 and C2 were detected in chorionic tissue and were secreted extracellularly. In chorion-derived cells, IL-1beta stimulated factor B synthesis but had no effect on C1r, C1 inhibitor, C1s, factor H and C4. TNFalpha had no stimulative effect on any of the complement proteins tested. In contrast, both IL-1beta and TNFalpha highly induced IL-6 secretion in chorion-derived cells, demonstrating the overall responsiveness of these cells to these stimuli. Interestingly, IFN-gamma increased the synthesis of C1s, C1r, C1 inhibitor, C4 and factor H in chorion-derived cells. The fact that the latter two complement proteins have opposing effects on immune activation of the complement cascade demonstrates the complex balance required to both maintain an ability to ward off infections but simultaneously suppress the immune response to enable tolerance of the allograft fetus.
Collapse
Affiliation(s)
- M Goldberg
- Institute of Allergy and Immunology, Assaf Harofeh Medical Center, Zerifin 70300, Israel.
| | | | | | | |
Collapse
|
45
|
Fairweather D, Frisancho-Kiss S, Njoku DB, Nyland JF, Kaya Z, Yusung SA, Davis SE, Frisancho JA, Barrett MA, Rose NR. Complement receptor 1 and 2 deficiency increases coxsackievirus B3-induced myocarditis, dilated cardiomyopathy, and heart failure by increasing macrophages, IL-1beta, and immune complex deposition in the heart. THE JOURNAL OF IMMUNOLOGY 2006; 176:3516-24. [PMID: 16517720 DOI: 10.4049/jimmunol.176.6.3516] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Complement and complement receptors (CR) play a central role in immune defense by initiating the rapid destruction of invading microorganisms, amplifying the innate and adaptive immune responses, and mediating solubilization and clearance of immune complexes. Defects in the expression of C or CR have been associated with loss of tolerance to self proteins and the development of immune complex-mediated autoimmune diseases such as systemic lupus erythematosus. In this study, we examined the role of CR on coxsackievirus B3 (CVB3)-induced myocarditis using mice deficient in CR1/2. We found that CR1/2 deficiency significantly increased acute CVB3 myocarditis and pericardial fibrosis resulting in early progression to dilated cardiomyopathy and heart failure. The increase in inflammation was not due to increased viral replication, which was not significantly altered in the hearts of CR1/2-deficient mice, but was associated with increased numbers of macrophages, IL-1beta levels, and immune complex deposition in the heart. The complement regulatory protein, CR1-related gene/protein Y (Crry), was increased on cardiac macrophage populations, while immature B220(low) B cells were increased in the spleen of CR1/2-deficient mice during acute CVB3-induced myocarditis. These results show that expression of CR1/2 is not necessary for effective clearance of CVB3 infection, but prevents immune-mediated damage to the heart.
Collapse
Affiliation(s)
- DeLisa Fairweather
- Department of Environmental Health Sciences, Johns Hopkins University and Bloomberg School of Public Health, 615 North Wolfe Street, Rm. E7628, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Fulya I, Mehmet O, Handan A, Vedat B. CYTOKINE MEASUREMENT IN LYMPHOCYTE CULTURE SUPERNATANT OF INACTIVE LEPROMATOUS LEPROSY PATIENTS. Indian J Med Microbiol 2006. [DOI: 10.1016/s0255-0857(21)02411-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
47
|
Baldwin L, Flanagan BF, Hunt JA. Flow cytometric measurement of phagocytosis reveals a role for C3b in metal particle uptake by phagocytes. J Biomed Mater Res A 2005; 73:80-5. [PMID: 15714502 DOI: 10.1002/jbm.a.30252] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A methodology for the quick and efficient study of phagocytosis has been developed. It uses the flow cytometer to exploit the change in size and granularity that occurs in cells upon the ingestion of particulate material. The numbers of cells that have phagocytosed particles can be calculated from the distinct shift in regions that occurs. The method also allows the factors governing phagocytosis to be studied in detail through the use of blocking agents or antibodies. Blood-derived monocytes were studied to investigate the role of complement in metal particle phagocytosis to further understand aseptic loosening. Factor C3b was found to be fundamental to the opsonization and phagocytosis of metal particles by monocytes.
Collapse
Affiliation(s)
- L Baldwin
- UKCTE, Clinical Engineering, University of Liverpool, L69 3GA, United Kingdom.
| | | | | |
Collapse
|
48
|
Hisano S, Matsushita M, Fujita T, Iwasaki H. Activation of the lectin complement pathway in Henoch-Schönlein purpura nephritis. Am J Kidney Dis 2005; 45:295-302. [DOI: 10.1053/j.ajkd.2004.10.020] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
49
|
Terwilliger DP, Clow LA, Gross PS, Smith LC. Constitutive expression and alternative splicing of the exons encoding SCRs in Sp152, the sea urchin homologue of complement factor B. Implications on the evolution of the Bf/C2 gene family. Immunogenetics 2004; 56:531-43. [PMID: 15448941 DOI: 10.1007/s00251-004-0711-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2004] [Revised: 07/14/2004] [Indexed: 01/26/2023]
Abstract
The purple sea urchin, Strongylocentrotus purpuratus, possesses a non-adaptive immune system including elements homologous to C3 and factor B (Bf) of the vertebrate complement system. SpBf is composed of motifs typical of the Bf/C2 protein family. Expression of Sp152 (encodes SpBf) was identified in the phagocyte type of coelomocyte in addition to gut, pharynx and esophagus, which may have been due to the presence of these coelomocytes in and on all tissues of the animal. Sp152 expression in coelomocytes was constitutive and non-inducible based on comparisons between pre- and post-injection with lipopolysaccharide or sterile seawater. The pattern of five short consensus repeats (SCRs) in SpBf has been considered ancestral compared to other deuterostome Bf/C2 proteins that contain either three or four SCRs. Three alternatively spliced messages were identified for Sp152 and designated Sp152Delta1, Sp152Delta4, and Sp152Delta1+Delta4, based on which of the five SCRs were deleted. Sp152Delta4 had an in-frame deletion of SCR4, which would encode a putative SpBfDelta4 protein with four SCRs rather than five. On the other hand, both Sp152Delta1 and Sp152Delta1+Delta4 had a frame-shift that introduced a stop codon six amino acids downstream of the splice site for SCR1, and would encode putative proteins composed only of the leader. Comparisons between the full-length SpBf and its several splice variants with other Bf/C2 proteins suggested that the early evolution of this gene family may have involved a combination of gene duplications and deletions of exons encoding SCRs.
Collapse
Affiliation(s)
- David P Terwilliger
- Department of Biological Sciences, The George Washington University, Lisner Hall 340, 2023 G Street, NW, Washington, DC 20052, USA
| | | | | | | |
Collapse
|
50
|
Kang TJ, Yeum CE, Kim BC, You EY, Chae GT. Differential production of interleukin-10 and interleukin-12 in mononuclear cells from leprosy patients with a Toll-like receptor 2 mutation. Immunology 2004; 112:674-80. [PMID: 15270740 PMCID: PMC1782533 DOI: 10.1111/j.1365-2567.2004.01926.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Toll-like receptor 2 (TLR2) is a key mediator of the immune response to mycobacterial infections, and mutations in TLR2 have been shown to confer susceptibility to infection with mycobacteria. This study investigated the profiles of cytokines, such as interferon (IFN)-gamma, interleukin (IL)-10, IL-12 and tumour necrosis factor (TNF)-alpha in response to Mycobacterium leprae in peripheral blood mononuclear cells (PBMC) with the TLR2 mutation Arg677Trp, a recently reported polymorphism that is associated with lepromatous leprosy. In leprosy patients with the TLR2 mutation, production of IL-2, IL-12, IFN-gamma, and TNF-alpha by M. leprae-stimulated PBMC were significantly decreased compared with that in groups with wild-type TLR2. However, the cells from patients with the TLR2 mutation showed significantly increased production of IL-10. There was no significant difference in IL-4 production between the mutant and wild-type during stimulation. Thus, these results suggest that the TLR2 signal pathway plays a critical role in the alteration of cytokine profiles in PBMC from leprosy patients and the TLR2 mutation Arg677Trp provides a mechanism for the poor cellular immune response associated with lepromatous leprosy.
Collapse
Affiliation(s)
- Tae Jin Kang
- Institute of Hansen's Disease, Department of Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-Dong, Seocho-Gu, Seoul 137-701, Republic of Korea
| | | | | | | | | |
Collapse
|