1
|
d'Amati A, Ronca R, Maccarinelli F, Turati M, Lorusso L, De Giorgis M, Tamma R, Ribatti D, Annese T. PTX3 shapes profibrotic immune cells and epithelial/fibroblast repair and regeneration in a murine model of pulmonary fibrosis. Pathol Res Pract 2023; 251:154901. [PMID: 37922722 DOI: 10.1016/j.prp.2023.154901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023]
Abstract
The long pentraxin 3 (PTX3) is protective in different pathologies but was not analyzed in-depth in Idiopathic Pulmonary Fibrosis (IPF). Here, we have explored the influence of PTX3 in the bleomycin (BLM)-induced murine model of IPF by looking at immune cells (macrophages, mast cells, T cells) and stemness/regenerative markers of lung epithelium (SOX2) and fibro-blasts/myofibroblasts (CD44) at different time points that retrace the progression of the disease from onset at day 14, to full-blown disease at day 21, to incomplete regression at day 28. We took advantage of transgenic PTX3 overexpressing mice (Tie2-PTX3) and Ptx3 null ones (PTX3-KO) in which pulmonary fibrosis was induced. Our data have shown that PTX3 overexpression in Tie2-PTX3 compared to WT or PTX3-KO: reduced CD68+ and CD163+ macrophages and the Tryptase+ mast cells during the whole experimental time; on the contrary, CD4+ T cells are consistently present on day 14 and dramatically decreased on day 21; CD8+ T cells do not show significant differences on day 14, but are significantly reduced on day 21; SOX2 is reduced on days 14 and 21; CD44 is reduced on day 21. Therefore, PTX3 could act on the proimmune and fibrogenic microenvironment to prevent fibrosis in BLM-treated mice.
Collapse
Affiliation(s)
- Antonio d'Amati
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy; Section of Pathology, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, 25123 Brescia, Italy
| | - Federica Maccarinelli
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, 25123 Brescia, Italy
| | - Marta Turati
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, 25123 Brescia, Italy
| | - Loredana Lorusso
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Michelina De Giorgis
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy; Department of Medicine and Surgery, LUM University, Casamassima, 70010 Bari, Italy.
| |
Collapse
|
2
|
Schmitz J, Kolaparambil Varghese LJ, Liebold F, Meyer M, Nerlich L, Starck C, Thierry S, Jansen S, Hinkelbein J. Influence of 30 and 60 Min of Hypobaric Hypoxia in Simulated Altitude of 15,000 ft on Human Proteome Profile. Int J Mol Sci 2022; 23:ijms23073909. [PMID: 35409267 PMCID: PMC8999033 DOI: 10.3390/ijms23073909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
The human body reacts to hypobaric hypoxia, e.g., during a stay at high altitude, with several mechanisms of adaption. Even short-time exposition to hypobaric hypoxia leads to complex adaptions. Proteomics facilitates the possibility to detect changes in metabolism due to changes in proteins. The present study aims to identify time-dependent changes in protein expression due to hypobaric hypoxia for 30 and 60 min at a simulated altitude of 15,000 ft. N = 80 male subjects were randomized and assigned into four different groups: 40 subjects to ground control for 30 (GC30) and 60 min (GC60) and 40 subjects to 15,000 ft for 30 (HH30) and 60 min (HH60). Subjects in HH30 and HH60 were exposed to hypobaric hypoxia in a pressure chamber (total pressure: 572 hPa) equivalent to 15,000 ft for 30 vs. 60 min, respectively. Drawn blood was centrifuged and plasma frozen (−80 °C) until proteomic analysis. After separation of high abundant proteins, protein expression was analyzed by 2-DIGE and MALDI-TOF. To visualize the connected signaling cascade, a bio-informatical network analysis was performed. The present study was approved by the ethical committee of the University of Cologne, Germany. The study registry number is NCT03823677. In comparing HH30 to GC30, a total of seven protein spots had a doubled expression, and 22 spots had decreased gene expression. In a comparison of HH60 to GC60, a total of 27 protein spots were significantly higher expressed. HH60, as compared to GC30, revealed that a total of 37 spots had doubled expression. Vice versa, 12 spots were detected, which were higher expressed in GC30 vs. HH60. In comparison to GC, HH60 had distinct differences in the number of differential protein spots (noticeably more proteins due to longer exposure to hypoxia). There are indicators that changes in proteins are dependent on the length of hypobaric hypoxia. Some proteins associated with hemostasis were differentially expressed in the 60 min comparison.
Collapse
Affiliation(s)
- Jan Schmitz
- Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.L.); (J.H.)
- German Society of Aerospace Medicine (DGLRM), 80331 Munich, Germany;
- Space Medicine Group, European Society of Aerospace Medicine (ESAM), 51149 Cologne, Germany;
- Department of Sleep and Human Factors Research, German Aerospace Center, Institute of Aerospace Medicine, 51147 Cologne, Germany
- Correspondence:
| | - Lydia J. Kolaparambil Varghese
- Space Medicine Group, European Society of Aerospace Medicine (ESAM), 51149 Cologne, Germany;
- Faculty of Medicine and Surgery, Università degli Studi di Perugia (Terni), 01500 Perugia, Italy
| | - Felix Liebold
- Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.L.); (J.H.)
- German Society of Aerospace Medicine (DGLRM), 80331 Munich, Germany;
- Space Medicine Group, European Society of Aerospace Medicine (ESAM), 51149 Cologne, Germany;
| | - Moritz Meyer
- Department of Otorhinolaryngology, Faculty of Medicine and University Hospital Essen, University of Essen, 45147 Essen, Germany;
| | - Lukas Nerlich
- German Society of Aerospace Medicine (DGLRM), 80331 Munich, Germany;
| | - Clement Starck
- Anesthesiology and Intensive Care Department, University Hospital of Brest, 29200 Brest, France;
| | - Seamus Thierry
- Anesthesiology Department, South Brittany General Hospital, 56322 Lorient, France;
| | - Stefanie Jansen
- Head and Neck Surgery, Department of Otorhinolaryngology, Medical Faculty, University of Cologne, 50937 Cologne, Germany;
| | - Jochen Hinkelbein
- Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.L.); (J.H.)
- German Society of Aerospace Medicine (DGLRM), 80331 Munich, Germany;
- Space Medicine Group, European Society of Aerospace Medicine (ESAM), 51149 Cologne, Germany;
| |
Collapse
|
3
|
Cockram TOJ, Dundee JM, Popescu AS, Brown GC. The Phagocytic Code Regulating Phagocytosis of Mammalian Cells. Front Immunol 2021; 12:629979. [PMID: 34177884 PMCID: PMC8220072 DOI: 10.3389/fimmu.2021.629979] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 05/18/2021] [Indexed: 01/21/2023] Open
Abstract
Mammalian phagocytes can phagocytose (i.e. eat) other mammalian cells in the body if they display certain signals, and this phagocytosis plays fundamental roles in development, cell turnover, tissue homeostasis and disease prevention. To phagocytose the correct cells, phagocytes must discriminate which cells to eat using a 'phagocytic code' - a set of over 50 known phagocytic signals determining whether a cell is eaten or not - comprising find-me signals, eat-me signals, don't-eat-me signals and opsonins. Most opsonins require binding to eat-me signals - for example, the opsonins galectin-3, calreticulin and C1q bind asialoglycan eat-me signals on target cells - to induce phagocytosis. Some proteins act as 'self-opsonins', while others are 'negative opsonins' or 'phagocyte suppressants', inhibiting phagocytosis. We review known phagocytic signals here, both established and novel, and how they integrate to regulate phagocytosis of several mammalian targets - including excess cells in development, senescent and aged cells, infected cells, cancer cells, dead or dying cells, cell debris and neuronal synapses. Understanding the phagocytic code, and how it goes wrong, may enable novel therapies for multiple pathologies with too much or too little phagocytosis, such as: infectious disease, cancer, neurodegeneration, psychiatric disease, cardiovascular disease, ageing and auto-immune disease.
Collapse
Affiliation(s)
| | | | | | - Guy C. Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Brilland B, Vinatier E, Subra JF, Jeannin P, Augusto JF, Delneste Y. Anti-Pentraxin Antibodies in Autoimmune Diseases: Bystanders or Pathophysiological Actors? Front Immunol 2021; 11:626343. [PMID: 33664737 PMCID: PMC7921723 DOI: 10.3389/fimmu.2020.626343] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/29/2020] [Indexed: 11/16/2022] Open
Abstract
Pentraxins are soluble innate immunity receptors involved in sensing danger molecules. They are classified as short (CRP, SAP) and long pentraxin subfamilies, including the prototypic long pentraxin PTX3. Pentraxins act mainly as bridging molecules favoring the clearance of microbes and dead cells. They are also involved in many other biological processes, such as regulation of complement activation, inflammation and tissue homeostasis. Autoantibodies directed against pentraxins have been reported in various autoimmune diseases, especially in systemic lupus erythematosus and ANCA-associated vasculitis. In this review, we review the main biological characteristics and functions of pentraxins and summarize data concerning autoantibodies directed against pentraxins in the context of autoimmune diseases and discuss their potential pathological role.
Collapse
Affiliation(s)
- Benoit Brilland
- CHU Angers, Service de Néphrologie-Dialyse-Transplantation, Angers, France.,Université d'Angers, INSERM, CRCINA, Angers, France
| | - Emeline Vinatier
- Université d'Angers, INSERM, CRCINA, Angers, France.,CHU Angers, Service d'Immunologie et Allergologie, Angers, France
| | - Jean-François Subra
- CHU Angers, Service de Néphrologie-Dialyse-Transplantation, Angers, France.,Université d'Angers, INSERM, CRCINA, Angers, France
| | - Pascale Jeannin
- Université d'Angers, INSERM, CRCINA, Angers, France.,CHU Angers, Service d'Immunologie et Allergologie, Angers, France
| | - Jean-François Augusto
- CHU Angers, Service de Néphrologie-Dialyse-Transplantation, Angers, France.,Université d'Angers, INSERM, CRCINA, Angers, France
| | - Yves Delneste
- Université d'Angers, INSERM, CRCINA, Angers, France.,CHU Angers, Service d'Immunologie et Allergologie, Angers, France
| |
Collapse
|
5
|
Wu Q, Cao F, Tao J, Li X, Zheng SG, Pan HF. Pentraxin 3: A promising therapeutic target for autoimmune diseases. Autoimmun Rev 2020; 19:102584. [PMID: 32534154 DOI: 10.1016/j.autrev.2020.102584] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/15/2022]
Abstract
Pentraxin 3 (PTX3) is a prototypic humoral soluble pattern recognition molecule that exerts a pivotal role in innate immune response and inflammation, as well as in tissue damage and remodeling. Recently, emerging evidence has revealed that PTX3 is involved in the occurrence and development of various autoimmune diseases, such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), ankylosing spondylitis (AS), systemic sclerosis (SSc), inflammatory bowel disease (IBD), multiple sclerosis (MS) and psoriasis, etc. In this review, we have succinctly summarized the complex immunological functions of PTX3 and mostly focused on recent findings of the pleiotropic activities played by PTX3 in the pathogenesis of autoimmune diseases, aiming at hopefully offering possible future therapeutic alternatives.
Collapse
Affiliation(s)
- Qian Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, 81 Meishan Road, Hefei, Anhui, China
| | - Fan Cao
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaomei Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui, China
| | - Song Guo Zheng
- Department of Internal Medicine, Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, 81 Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
6
|
Ramirez GA, Rovere-Querini P, Blasi M, Sartorelli S, Di Chio MC, Baldini M, De Lorenzo R, Bozzolo EP, Leone R, Mantovani A, Manfredi AA, Tombetti E. PTX3 Intercepts Vascular Inflammation in Systemic Immune-Mediated Diseases. Front Immunol 2019; 10:1135. [PMID: 31191526 PMCID: PMC6548810 DOI: 10.3389/fimmu.2019.01135] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/07/2019] [Indexed: 01/02/2023] Open
Abstract
PTX3 is a prototypic soluble pattern recognition receptor, expressed at sites of inflammation and involved in regulation of the tissue homeostasis. PTX3 systemic levels increase in many (but not all) immune-mediated inflammatory conditions. Research on PTX3 as a biomarker has so far focused on single diseases. Here, we performed a multi-group comparative study with the aim of identifying clinical and pathophysiological phenotypes associated with PTX3 release. PTX3 concentration was measured by ELISA in the plasma of 366 subjects, including 96 patients with giant cell arteritis (GCA), 42 with Takayasu's arteritis (TA), 10 with polymyalgia rheumatica (PMR), 63 with ANCA-associated systemic small vessel vasculitides (AAV), 55 with systemic lupus erythematosus (SLE), 21 with rheumatoid arthritis (RA) and 79 healthy controls (HC). Patients with SLE, AAV, TA and GCA, but not patients with RA and PMR, had higher PTX3 levels than HC. PTX3 concentration correlated with disease activity, acute phase reactants and prednisone dose. It was higher in females, in patients with recent-onset disease and in those with previous or current active vasculitis at univariate analysis. Active small- or large- vessel vasculitis were the main independent variables influencing PTX3 levels at multivariate analysis. High levels of PTX3 in the blood can contribute to identify an increased risk of vascular involvement in patients with systemic immune-mediated diseases.
Collapse
Affiliation(s)
- Giuseppe A. Ramirez
- Università Vita-Salute San Raffaele, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Patrizia Rovere-Querini
- Università Vita-Salute San Raffaele, Milan, Italy
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Miriam Blasi
- Università Vita-Salute San Raffaele, Milan, Italy
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | | | - Mattia Baldini
- Università Vita-Salute San Raffaele, Milan, Italy
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Rebecca De Lorenzo
- Università Vita-Salute San Raffaele, Milan, Italy
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Enrica P. Bozzolo
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Alberto Mantovani
- Humanitas Research Center - IRCCS, Rozzano, Italy
- Humanitas University, Rozzano, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Angelo A. Manfredi
- Università Vita-Salute San Raffaele, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Enrico Tombetti
- Università Vita-Salute San Raffaele, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
7
|
Erreni M, Manfredi AA, Garlanda C, Mantovani A, Rovere-Querini P. The long pentraxin PTX3: A prototypical sensor of tissue injury and a regulator of homeostasis. Immunol Rev 2018; 280:112-125. [PMID: 29027216 DOI: 10.1111/imr.12570] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tissue damage frequently occurs. The immune system senses it and enforces homeostatic responses that lead to regeneration and repair. The synthesis of acute phase molecules is emerging as a crucial event in this program. The prototypic long pentraxin PTX3 orchestrates the recruitment of leukocytes, stabilizes the provisional matrix in order to facilitate leukocyte and stem progenitor cells trafficking, promotes swift and safe clearance of dying cells and of autoantigens, limiting autoimmunity and protecting the vasculature. These non-redundant actions of PTX3 are necessary for the resolution of inflammation. Recent studies have highlighted the mechanisms by which PTX3 adapts the functions of innate immune cells, orchestrates tissue repair and contributes to select the appropriate acquired immune response in various tissues. Conversely, PTX3 continues to be produced in diseases where the inflammatory response does not resolve. It is therefore a valuable biomarker for more precise and personalized stratification of patients, often independently predicting clinical evolution and outcome. There is strong promise for novel therapies based on understanding the mechanisms with which PTX3 plays its homeostatic role, especially in regulating leukocyte migration and the resolution of inflammatory processes.
Collapse
Affiliation(s)
- Marco Erreni
- IRCCS Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy
| | - Angelo A Manfredi
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy
| | - Alberto Mantovani
- IRCCS Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy
| | - Patrizia Rovere-Querini
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
8
|
Zhang D, Xi M, Chen L, Huang Y, Mao P. PTX3 in serum induces renal mesangial cell proliferation but has no effect on apoptosis. Exp Ther Med 2018; 15:1193-1198. [PMID: 29434706 PMCID: PMC5774436 DOI: 10.3892/etm.2017.5521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 04/21/2017] [Indexed: 12/22/2022] Open
Abstract
The present study aimed to investigate the effect of pentraxin 3 (PTX3) on the regulation of proliferation and apoptosis in human glomerular mesangial cells (HMCs). Small interfering (si)RNA was designed and synthesized to inhibit the expression of endogenous PTX3, and the effects on the proliferation and apoptosis of HMCs were detected by flow cytometry and an MTT assay. Western blot analysis was used to detect the activation of mitogen-activated protein kinase (MAPK) proteins in HMCs with PTX3 knockdown. Three siRNAs targeting PTX3 were individually transfected into HMCs for 48 h, and reverse-transcription quantitative PCR demonstrated that the relative mRNA expression of PTX3 was significantly decreased in all groups by up to 79.62% of that in the control group (P<0.05). Following transfection with PTX3-siRNA, the viability of an HMC line was significantly decreased in comparison with that of a control group transfected with scrambled siRNA. However, PTX3-siRNA did not significantly effect early and late apoptotic cell populations in HMCs compared with those in the control. Endogenous PTX3 interference was found to significantly decrease p38 MAPK, extracellular signal-regulated kinase 1/2 and c-Jun N-terminal kinase phosphorylation. In conclusion, silencing of PTX3, inhibited the proliferation of HMCs via MAPK pathways, but exerted no effect on the apoptosis of HMCs.
Collapse
Affiliation(s)
- Danhuan Zhang
- Department of Nephrology, Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200336, P.R. China
| | - Minhui Xi
- Department of Nephrology, Pudong New Area People's Hospital, Shanghai 201200, P.R. China
| | - Lingyun Chen
- Department of Nephrology, Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200336, P.R. China
| | - Yanping Huang
- Department of Nephrology, Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200336, P.R. China
| | - Peiju Mao
- Department of Nephrology, Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200336, P.R. China
| |
Collapse
|
9
|
Assandri R, Montanelli A. Pentraxin 3 and biopsy status in celiac patients. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2018; 11:225-232. [PMID: 30013746 PMCID: PMC6040031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AIM In our study we explored a possible relationship between PTX3 and CD. BACKGROUND Gluten sensitivity is known as a hallmark of celiac disease (CD). The diagnosis of CD requires demonstration of a typical enteropathy, and positive serology supports the diagnosis. The CD immune response involves the adaptive, as well as the innate immunity and is characterized by the presence of anti-gliadin (AGA) and anti-transglutaminase 2 antibodies (tTGA), lymphocytic infiltration in the intestinal epithelial membrane and expression of multiple cytokines. The long pentraxin 3 (PTX3), an acute-phase inflammatory molecule, plays an important role in innate immunity. METHODS 108 CD patients were divided according to Marsh Histological grade following Marsh criteria classification in three groups: Group 1: Marsh 0, patients with a known history of CD under gluten free diet, complete remission; Group 2: Marsh1 and Marsh 2; Group 3: Marsh 3. Healthy age-matched controls without a known history of CD or gastrointestinal symptoms (n=30) served as controls. PTX3 serum levels were measured by sandwich ELISA on an automated platform. RESULTS PTX3 serum levels were significantly elevated in group 3 and group 2 compared with HC (mean 3.31± 1.27 ng/mL and 3.97 ± 0.54 ng/mL versus 1.06 ± 0.59 ng/mL; P < 0.005), with group 1 (0.76±0.31 ng/mL). No statistically significant differences were found between group 1 and HC group. We found a strong linear correlation between PTX3 serum levels and AGA levels in group 2 (r=0.78, P <0.0001), and group 3 (r =0.63, P < 0.005) but no correlations were detected between PTX3 serum levels and tTGA levels (group 2, r= 0.04; group 3, r=0.24). Serological data revealed that PTX3 correlated with major gastrointestinal damage patients. CONCLUSION PTX3 is a component of the humoral arm of the innate immune system. Our data showed that PTX3 serum levels were high in active disease patients with pathological levels of AGA. We also demonstrated that patients with normal AGA IgA levels had PTX3 serum levels compared to healthy control. We hypothesized that PTX3 is able to modulate the innate response to gliadin in CD and it could regulate the adaptive immune response.
Collapse
Affiliation(s)
- Roberto Assandri
- Departement of Clinical Pathology, Clinical Chemistry Laboratory ASST Ospedale Maggiore di Crema, Italy
| | | |
Collapse
|
10
|
Daigo K, Inforzato A, Barajon I, Garlanda C, Bottazzi B, Meri S, Mantovani A. Pentraxins in the activation and regulation of innate immunity. Immunol Rev 2017; 274:202-217. [PMID: 27782337 DOI: 10.1111/imr.12476] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Humoral fluid phase pattern recognition molecules (PRMs) are a key component of the activation and regulation of innate immunity. Humoral PRMs are diverse. We focused on the long pentraxin PTX3 as a paradigmatic example of fluid phase PRMs. PTX3 acts as a functional ancestor of antibodies and plays a non-redundant role in resistance against selected microbes in mouse and man and in the regulation of inflammation. This molecule interacts with complement components, thus modulating complement activation. In particular, PTX3 regulates complement-driven macrophage-mediated tumor progression, acting as an extrinsic oncosuppressor in preclinical models and selected human tumors. Evidence collected over the years suggests that PTX3 is a biomarker and potential therapeutic agent in humans, and pave the way to translation of this molecule into the clinic.
Collapse
Affiliation(s)
- Kenji Daigo
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Antonio Inforzato
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano (Milan), Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Italy
| | | | - Cecilia Garlanda
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Barbara Bottazzi
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Seppo Meri
- Immunobiology Research Program, Research Programs Unit, Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki , Helsinki , Finland
| | - Alberto Mantovani
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano (Milan), Italy.,Humanitas University, Rozzano, Italy
| |
Collapse
|
11
|
Monocyte differentiation and macrophage priming are regulated differentially by pentraxins and their ligands. BMC Immunol 2017; 18:30. [PMID: 28619036 PMCID: PMC5472910 DOI: 10.1186/s12865-017-0214-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/08/2017] [Indexed: 01/01/2023] Open
Abstract
Background Circulating bone marrow-derived monocytes can leave the blood, enter a tissue, and differentiate into M1 inflammatory, M2a remodeling/fibrotic, or M2c/Mreg resolving/immune-regulatory macrophages. Macrophages can also convert from one of the above types to another. Pentraxins are secreted proteins that bind to, and promote efficient clearance of, microbial pathogens and cellular debris during infection, inflammation, and tissue damage. The pentraxins C-reactive protein (CRP), serum amyloid P (SAP), and pentraxin-3 (PTX3) can also bind a variety of endogenous ligands. As monocytes and macrophages are exposed to differing concentrations of pentraxins and their ligands during infection, inflammation, and tissue damage, we assessed what effect pentraxins and their ligands have on these cells. Results We found that many polarization markers do not discriminate between the effects of pentraxins and their ligands on macrophages. However, pentraxins, their ligands, and cytokines differentially regulate the expression of the hemoglobin-haptoglobin complex receptor CD163, the sialic acid-binding lectin CD169, and the macrophage mannose receptor CD206. CRP, a pentraxin generally thought of as being pro-inflammatory, increases the extracellular accumulation of the anti-inflammatory cytokine IL-10, and this effect is attenuated by GM-CSF, mannose-binding lectin, and factor H. Conclusions These results suggest that the presence of pentraxins and their ligands regulate macrophage differentiation in the blood and tissues, and that CRP may be a potent inducer of the anti-inflammatory cytokine IL-10. Electronic supplementary material The online version of this article (doi:10.1186/s12865-017-0214-z) contains supplementary material, which is available to authorized users.
Collapse
|
12
|
Augusto JF, Poli C, Beauvillain C, Subra JF, Jaillon S, Renier G, Chevailler A, Puéchal X, Delneste Y, Jeannin P. Anti-pentraxin antibodies in autoimmune systemic diseases: Focus on anti-pentraxin-3 autoantibodies. Int Rev Immunol 2017; 36:145-153. [DOI: 10.1080/08830185.2017.1284210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jean-François Augusto
- Department of Nephrology-Dialysis-Transplantation, University Hospital of Angers, Angers, France
- Angers University Hospital, University of Angers, Angers, France
- CRCINA, INSERM, Université de Nantes, Université d'Angers, LabEx IGO “Immunotherapy, Graft, Oncology”, Angers, France
| | - Caroline Poli
- Angers University Hospital, University of Angers, Angers, France
| | - Céline Beauvillain
- Angers University Hospital, University of Angers, Angers, France
- Immunology and Allergology Laboratory, University Hospital of Angers, Angers, France
- CRCINA, INSERM, Université de Nantes, Université d'Angers, LabEx IGO “Immunotherapy, Graft, Oncology”, Angers, France
| | - Jean-François Subra
- Department of Nephrology-Dialysis-Transplantation, University Hospital of Angers, Angers, France
- Angers University Hospital, University of Angers, Angers, France
- CRCINA, INSERM, Université de Nantes, Université d'Angers, LabEx IGO “Immunotherapy, Graft, Oncology”, Angers, France
| | - Sebastien Jaillon
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
| | - Gilles Renier
- Immunology and Allergology Laboratory, University Hospital of Angers, Angers, France
| | - Alain Chevailler
- Angers University Hospital, University of Angers, Angers, France
- Immunology and Allergology Laboratory, University Hospital of Angers, Angers, France
- CRCINA, INSERM, Université de Nantes, Université d'Angers, LabEx IGO “Immunotherapy, Graft, Oncology”, Angers, France
| | - Xavier Puéchal
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Yves Delneste
- Angers University Hospital, University of Angers, Angers, France
- Immunology and Allergology Laboratory, University Hospital of Angers, Angers, France
- CRCINA, INSERM, Université de Nantes, Université d'Angers, LabEx IGO “Immunotherapy, Graft, Oncology”, Angers, France
| | - Pascale Jeannin
- Angers University Hospital, University of Angers, Angers, France
- Immunology and Allergology Laboratory, University Hospital of Angers, Angers, France
- CRCINA, INSERM, Université de Nantes, Université d'Angers, LabEx IGO “Immunotherapy, Graft, Oncology”, Angers, France
| |
Collapse
|
13
|
Vezzoli M, Sciorati C, Campana L, Monno A, Doglio MG, Rigamonti E, Corna G, Touvier T, Castiglioni A, Capobianco A, Mantovani A, Manfredi AA, Garlanda C, Rovere-Querini P. The clearance of cell remnants and the regeneration of the injured muscle depend on soluble pattern recognition receptor PTX3. Mol Med 2016; 22:809-820. [PMID: 27900389 DOI: 10.2119/molmed.2016.00002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 09/29/2016] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE The signals causing the resolution of muscle inflammation are only partially characterized. The long pentraxin PTX3, which modulates leukocyte recruitment and activation, could contribute. METHODS We analysed the expression of ptx3 after muscle injury and verified whether hematopoietic precursors are a source of the protein. The kinetics of regeneration and leukocytes infiltration, the accumulation of cell remnants and anti-histidyl-t-RNA synthetase autoantibodies were compared in wild-type and ptx3-deficient mice. RESULTS Ptx3 expression was up-regulated three-five days after injury and restricted to the extracellular matrix. Cellular debris and leukocytes persisted in the muscle of ptx3-deficient mice for a long time after wild-type animals had healed. ptx3-deficient macrophages expressed receptors involved in apoptotic cell clearance and engulfed dead cells in vitro. Accumulation of cell debris in a pro-inflammatory microenvironment was not sufficient to elicit autoantibodies. CONCLUSION PTX3 generated in response to muscle injury prompts the clearance of debris and the termination of the inflammatory response.
Collapse
Affiliation(s)
- Michela Vezzoli
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy
| | - Clara Sciorati
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy
| | - Lara Campana
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy
| | - Antonella Monno
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy
| | - Maria Giulia Doglio
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy
| | - Elena Rigamonti
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy
| | - Gianfranca Corna
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy
| | - Thierry Touvier
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy
| | - Alessandra Castiglioni
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy
| | - Annalisa Capobianco
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Rozzano, Milano, Italy.,Humanitas University, Rozzano, Milan, Italy
| | - Angelo A Manfredi
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy.,Vita-Salute San Raffaele University, School of Medicine, Milano, Italy
| | | | - Patrizia Rovere-Querini
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milano, Italy.,Vita-Salute San Raffaele University, School of Medicine, Milano, Italy
| |
Collapse
|
14
|
The Immune System in Tissue Environments Regaining Homeostasis after Injury: Is "Inflammation" Always Inflammation? Mediators Inflamm 2016; 2016:2856213. [PMID: 27597803 PMCID: PMC4997018 DOI: 10.1155/2016/2856213] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 07/08/2016] [Accepted: 07/19/2016] [Indexed: 12/31/2022] Open
Abstract
Inflammation is a response to infections or tissue injuries. Inflammation was once defined by clinical signs, later by the presence of leukocytes, and nowadays by expression of "proinflammatory" cytokines and chemokines. But leukocytes and cytokines often have rather anti-inflammatory, proregenerative, and homeostatic effects. Is there a need to redefine "inflammation"? In this review, we discuss the functions of "inflammatory" mediators/regulators of the innate immune system that determine tissue environments to fulfill the need of the tissue while regaining homeostasis after injury.
Collapse
|
15
|
Mistry P, Kaplan MJ. Cell death in the pathogenesis of systemic lupus erythematosus and lupus nephritis. Clin Immunol 2016; 185:59-73. [PMID: 27519955 DOI: 10.1016/j.clim.2016.08.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 12/12/2022]
Abstract
Nephritis is one of the most severe complications of systemic lupus erythematosus (SLE). One key characteristic of lupus nephritis (LN) is the deposition of immune complexes containing nucleic acids and/or proteins binding to nucleic acids and autoantibodies recognizing these molecules. A variety of cell death processes are implicated in the generation and externalization of modified nuclear autoantigens and in the development of LN. Among these processes, apoptosis, primary and secondary necrosis, NETosis, necroptosis, pyroptosis, and autophagy have been proposed to play roles in tissue damage and immune dysregulation. Cell death occurs in healthy individuals during conditions of homeostasis yet autoimmunity does not develop, at least in part, because of rapid clearance of dying cells. In SLE, accelerated cell death combined with a clearance deficiency may lead to the accumulation and externalization of nuclear autoantigens and to autoantibody production. In addition, specific types of cell death may modify autoantigens and alter their immunogenicity. These modified molecules may then become novel targets of the immune system and promote autoimmune responses in predisposed hosts. In this review, we examine various cell death pathways and discuss how enhanced cell death, impaired clearance, and post-translational modifications of proteins could contribute to the development of lupus nephritis.
Collapse
Affiliation(s)
- Pragnesh Mistry
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Balhara J, Shan L, Zhang J, Muhuri A, Halayko AJ, Almiski MS, Doeing D, McConville J, Matzuk MM, Gounni AS. Pentraxin 3 deletion aggravates allergic inflammation through a T H17-dominant phenotype and enhanced CD4 T-cell survival. J Allergy Clin Immunol 2016; 139:950-963.e9. [PMID: 27567326 PMCID: PMC6317853 DOI: 10.1016/j.jaci.2016.04.063] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 04/01/2016] [Accepted: 04/28/2016] [Indexed: 12/17/2022]
Abstract
Background Pentraxin 3 (PTX3) is a multifunctional molecule that plays a nonredundant role at the crossroads between pathogen clearance, innate immune system, matrix deposition, female fertility, and vascular biology. It is produced at sites of infection and inflammation by both structural and inflammatory cells. However, its role in allergen-induced inflammation remains to be tested. Objective We sought to determine the effect of Ptx3 deletion on ovalbumin (OVA)–induced allergic inflammation in a murine model of asthma. Methods Bronchoalveolar lavage fluid was collected from patients with severe asthma and healthy subjects, and the level of PTX3 was determined by using ELISA. Ptx3+/+ and Ptx3−/− mice were sensitized and challenged with OVA and bronchoalveolar lavage fluid, and the lungs were collected for assessing inflammation. Lung tissue inflammation and mucus production were assessed by means of flow cytometry and hematoxylin and eosin and periodic acid-Schiff staining, respectively. flexiVent was used to determine airway resistance to methacholine in these mice. Results Here we report that mice with severe asthma and OVA-sensitized/challenged mice had increased PTX3 levels in the lungs compared with healthy control mice. Mice lacking PTX3 have exaggerated neutrophilic/eosinophilic lung inflammation, mucus production, and airway hyperresponsiveness in an experimental model of OVA-induced asthma. Furthermore, OVA-exposed lung Ptx3−/− CD4 T cells exhibit an increased production of IL-17A, an effect that is accompanied by an increased signal transducer and activator of transcription 3 phosphorylation, reduced IL-2 production, and enhanced activation and survival. Also, we observed an increase in numbers of IL-6– and IL-23–producing dendritic cells in OVA-exposed Ptx3−/− mice compared with those in wild-type control mice. Conclusion Altogether, PTX3 deficiency results in augmented airway hyperresponsiveness, mucus production, and IL-17A–dominant pulmonary inflammation, suggesting a regulatory role of PTX3 in the development of allergic inflammation.
Collapse
Affiliation(s)
- Jyoti Balhara
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lianyu Shan
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jingbo Zhang
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anik Muhuri
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew J Halayko
- Department of Physiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Muhamad S Almiski
- Department of Pathology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Diana Doeing
- Department of Medicine, University of Chicago, Chicago, Ill
| | | | - Martin M Matzuk
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Tex
| | - Abdelilah S Gounni
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
17
|
Doehn JM, Winkler A, Kuzmina Z, Hladik A, Greinix H, Knapp S, Robak O. Pentraxin-3 levels in graft-versus-host disease during allogeneic hematopoietic stem cell transplantation. Exp Hematol 2016; 44:917-23. [PMID: 27403536 DOI: 10.1016/j.exphem.2016.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/23/2016] [Accepted: 06/29/2016] [Indexed: 01/15/2023]
Abstract
Acute and chronic graft-versus-host-diseases (aGVHD and cGVHD, respectively) are serious complications after hematopoietic stem cell transplantation (HSCT), impairing survival and quality of life. Because the underlying pathomechanism of GVHD is still poorly understood, we investigated the novel inflammatory marker Pentraxin-3 (PTX3) for its potential role in acute and chronic GVHD compared with autologous HSCT and healthy individuals. We collected plasma samples from patients undergoing autologous (n = 12) and allogeneic (n = 28) HSCT and from healthy individuals (n = 15) throughout 7 days before and up to 1 year after HSCT. PTX3 levels in patients with aGVHD were significantly higher (36.4 ± 23.6 ng/mL) than in allogeneic patients without aGVHD (10.4 ± 4.4 ng/mL, p = 0.0001), autologous controls (11.4 ± 6.7 ng/mL, p = 0.001), or healthy individuals (1.9 ± 0.6 ng/mL, p < 0.001). PTX3 levels in patients with cGVHD (13.6 ± 6.3 ng/mL) were significantly lower than in allogeneic patients without cGVHD (25.1 ± 13.8 ng/mL, p = 0.04) and higher than in autologous controls (8.9 ± 7.8 ng/mL, p = 0.07) and healthy individuals (1.9 ± 0.6 ng/mL, p < 0.001). Severity of aGVHD and cGVHD correlated with PTX3 levels. Rising PTX3 levels after HSCT indicated unfavorable outcome. We show that PTX3 levels correlate with the severity of aGVHD, cGVHD, and-with reservations-survival in patients undergoing allogeneic HSCT.
Collapse
Affiliation(s)
- Jan-Moritz Doehn
- Department of Infectious Diseases and Pneumology, Charité University Medicine Berlin, Berlin, Germany
| | - Andreas Winkler
- Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Zoya Kuzmina
- Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anastasiya Hladik
- Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Hildegard Greinix
- Department of Medicine I, Medical University of Vienna, Vienna, Austria; Division of Hematology, Medical University of Graz, Graz, Austria
| | - Sylvia Knapp
- Department of Medicine I, Medical University of Vienna, Vienna, Austria; CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Oliver Robak
- Department of Infectious Diseases and Pneumology, Charité University Medicine Berlin, Berlin, Germany; Department of Medicine I, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
18
|
Ketter P, Yu JJ, Cap AP, Forsthuber T, Arulanandam B. Pentraxin 3: an immune modulator of infection and useful marker for disease severity assessment in sepsis. Expert Rev Clin Immunol 2016; 12:501-7. [PMID: 26982005 DOI: 10.1586/1744666x.2016.1166957] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The acute phase protein pentraxin 3 (PTX3) is a pattern recognition receptor involved in regulation of the host immune response. This relatively newly discovered member of the pentraxin superfamily elicits both immunostimulatory and immunoregulatory functions preventing autoimmune pathology and orchestrated clearance of pathogens through opsonization of damage- and pathogen-associated molecular patterns (DAMP/PAMP). Thus, PTX3 has been described as a possible evolutionary precursor to immunoglobulins. While shown to provide protection against specific bacterial and fungal pathogens, persistent elevation of PTX3 levels following initial onset of infection appear to predict poor patient outcome and may contribute to disease sequelae such as tissue damage and coagulopathy. Measurement of PTX3 following onset of sepsis may improve patient risk assessment and thus be useful in guiding subsequent therapeutic interventions including steroidal anti-inflammatory and altered antibiotic therapies. In this review, we summarize the role of PTX3 in inflammatory syndromes and its utility as a marker of sepsis disease severity.
Collapse
Affiliation(s)
- Patrick Ketter
- a Blood and Coagulation Program , United States Army Institute of Surgical Research , JBSA-Fort Sam Houston , TX , USA
| | - Jieh-Juen Yu
- b Department of Biology , University of Texas at San Antonio , San Antonio , TX , USA
| | - Andrew P Cap
- a Blood and Coagulation Program , United States Army Institute of Surgical Research , JBSA-Fort Sam Houston , TX , USA
| | - Thomas Forsthuber
- b Department of Biology , University of Texas at San Antonio , San Antonio , TX , USA
| | - Bernard Arulanandam
- b Department of Biology , University of Texas at San Antonio , San Antonio , TX , USA
| |
Collapse
|
19
|
Pentraxin 3 Plasma Levels and Disease Activity in Systemic Lupus Erythematosus. Autoimmune Dis 2015; 2015:354014. [PMID: 26613049 PMCID: PMC4646995 DOI: 10.1155/2015/354014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 09/11/2015] [Accepted: 09/15/2015] [Indexed: 12/22/2022] Open
Abstract
SLE is an autoimmune disorder that involves polyclonal autoimmunity against multiple autoantigens. PTX3, a marker of the acute-phase inflammatory response, plays an important role in innate immunity and in modulation of the adaptive immune response. Our study tried to resolve some rather controversial aspects of the use of PTX3 as a biomarker of disease activity in SLE patients. We demonstrated that plasma PTX3 concentration of the SLE patients was significantly higher than the healthy control groups and reflected disease activity. ROC curve analysis was used to determine best cut-off point (2.8 ng/mL) with a good sensitivity and specificity. In patients with SLE, PTX3 concentrations were correlated with SLEDAI. Trend to remission (TTR) curve was created by plotting PTX3 levels and SLEDAI and we applied the curve as a model for the analysis of two patients with different follow-up. PTX3 plasma levels declined significantly and this decline occurred parallel to the clinical improvement with a complete remission of disease. In patients who experienced a clinical relapse, an increase in PTX3 levels followed the lupus flare. The proposal of PTX3 cut-off associated with TTR and monitoring of PTX3 plasma levels could be an innovative approach to follow-up of SLE patients.
Collapse
|
20
|
Dickerson F, Stallings C, Origoni A, Katsafanas E, Schweinfurth LAB, Savage CLG, Khushalani S, Yolken R. Pentraxin 3 is reduced in bipolar disorder. Bipolar Disord 2015; 17:409-14. [PMID: 25425421 DOI: 10.1111/bdi.12281] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/29/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Immunologic abnormalities have been found in bipolar disorder but pentraxin 3, a marker of innate immunity, has not been studied in this population. METHODS Levels of pentraxin 3 were measured in individuals with bipolar disorder, schizophrenia, and non-psychiatric controls. Linear regression models were used to compare the pentraxin 3 levels in each of the psychiatric groups to that in the control group, adjusting for demographic and clinical variables. Logistic regression models were used to calculate the odds ratios associated with levels of pentraxin 3 which differed from specified levels of the control group. RESULTS The sample consisted of 831 individuals: 256 with bipolar disorder, 309 with schizophrenia, and 266 without a psychiatric disorder. The levels of pentraxin 3 in the bipolar disorder, but not in the schizophrenia, group were significantly lower than those of controls, adjusting for age, gender, race, maternal education, smoking status, and body mass index (t = -3.78, p < 0.001). The individuals with bipolar disorder also had significantly increased odds of having low levels of pentraxin 3 relative to both the 10th and 25th percentile level of the controls and significantly decreased odds of having a level greater than the 75th and the 90th percentile level of the controls, adjusting for the same covariates. CONCLUSIONS Individuals with bipolar disorder have low levels of pentraxin 3 which may reflect impaired innate immunity. An increased understanding of the role of innate immunity in the etiopathogenesis of bipolar disorder might lead to new modalities for the diagnosis and treatment of this disorder.
Collapse
Affiliation(s)
- Faith Dickerson
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Cassie Stallings
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Andrea Origoni
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Emily Katsafanas
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | | | | | - Sunil Khushalani
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Robert Yolken
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Plasma pentraxin-3 and coagulation and fibrinolysis variables during acute Puumala hantavirus infection and associated thrombocytopenia. Blood Coagul Fibrinolysis 2015; 25:612-7. [PMID: 24751477 DOI: 10.1097/mbc.0000000000000117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Thrombocytopenia and altered coagulation characterize all hantavirus infections. To further assess the newly discovered predictive biomarkers of disease severity during acute Puumala virus (PUUV) infection, we studied the associations between them and the variables reflecting coagulation, fibrinolysis and endothelial activation. Nineteen hospital-treated patients with serologically confirmed acute PUUV infection were included. Acutely, plasma levels of pentraxin-3 (PTX3), cell-free DNA (cf-DNA), complement components SC5b-9 and C3 and interleukin-6 (IL-6) were recorded as well as platelet ligands and markers of coagulation and fibrinolysis. High values of plasma PTX3 associated with thrombin formation (prothrombin fragments F1+2; r = 0.46, P = 0.05), consumption of platelet ligand fibrinogen (r = -0.70, P < 0.001) and natural anticoagulants antithrombin (AT) (r = -0.74, P < 0.001), protein C (r = -0.77, P < 0.001) and protein S free antigen (r = -0.81, P < 0.001) and a decreased endothelial marker ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 domain 13) (r = -0.48, P = 0.04). Plasma level of AT associated with C3 (r = 0.76, P < 0.001), IL-6 (r = -0.56, P = 0.01) and cf-DNA (r = -0.47, P = 0.04). High cf-DNA coincided with increased prothrombin fragments F1+2 (r = 0.47, P = 0.04). Low C3 levels reflecting the activation of complement system through the alternative route predicted loss of all natural anticoagulants (for protein C r = 0.53, P = 0.03 and for protein S free antigen r = 0.64, P = 0.004). Variables depicting altered coagulation follow the new predictive biomarkers of disease severity, especially PTX3, in acute PUUV infection. The findings are consistent with the previous observations of these biomarkers also being predictive for low platelet count and underline the cross-talk of inflammation and coagulation systems in acute PUUV infection.
Collapse
|
22
|
Liao X, Ren J, Wei CH, Ross AC, Cecere TE, Jortner BS, Ahmed SA, Luo XM. Paradoxical effects of all-trans-retinoic acid on lupus-like disease in the MRL/lpr mouse model. PLoS One 2015; 10:e0118176. [PMID: 25775135 PMCID: PMC4361690 DOI: 10.1371/journal.pone.0118176] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 01/05/2015] [Indexed: 11/27/2022] Open
Abstract
Roles of all-trans-retinoic acid (tRA), a metabolite of vitamin A (VA), in both tolerogenic and immunogenic responses are documented. However, how tRA affects the development of systemic autoimmunity is poorly understood. Here we demonstrate that tRA have paradoxical effects on the development of autoimmune lupus in the MRL/lpr mouse model. We administered, orally, tRA or VA mixed with 10% of tRA (referred to as VARA) to female mice starting from 6 weeks of age. At this age, the mice do not exhibit overt clinical signs of lupus. However, the immunogenic environment preceding disease onset has been established as evidenced by an increase of total IgM/IgG in the plasma and expansion of lymphocytes and dendritic cells in secondary lymphoid organs. After 8 weeks of tRA, but not VARA treatment, significantly higher pathological scores in the skin, brain and lung were observed. These were accompanied by a marked increase in B-cell responses that included autoantibody production and enhanced expression of plasma cell-promoting cytokines. Paradoxically, the number of lymphocytes in the mesenteric lymph node decreased with tRA that led to significantly reduced lymphadenopathy. In addition, tRA differentially affected renal pathology, increasing leukocyte infiltration of renal tubulointerstitium while restoring the size of glomeruli in the kidney cortex. In contrast, minimal induction of inflammation with tRA in the absence of an immunogenic environment in the control mice was observed. Altogether, our results suggest that under a predisposed immunogenic environment in autoimmune lupus, tRA may decrease inflammation in some organs while generating more severe disease in others.
Collapse
Affiliation(s)
- Xiaofeng Liao
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, United States of America
| | - Jingjing Ren
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, United States of America
| | - Cheng-Hsin Wei
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, 16802, United States of America
| | - A. Catharine Ross
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, 16802, United States of America
| | - Thomas E. Cecere
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, United States of America
| | - Bernard S. Jortner
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, United States of America
| | - S. Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, United States of America
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, United States of America
- * E-mail:
| |
Collapse
|
23
|
Tan S, Gan C, Li R, Ye Y, Zhang S, Wu X, Yang YY, Fan W, Wu M. A novel chemosynthetic peptide with β-sheet motif efficiently kills Klebsiella pneumoniae in a mouse model. Int J Nanomedicine 2015; 10:1045-59. [PMID: 25709431 PMCID: PMC4330034 DOI: 10.2147/ijn.s73303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Klebsiella pneumoniae (Kp) is one of the most common pathogens in nosocomial infections and is increasingly becoming multiple drug resistant. However, the molecular pathogenesis of Kp in causing tissue injury and dysregulated host defense remains elusive, further dampening the development of novel therapeutic measures. We have previously screened a series of synthetic antimicrobial beta-sheet forming peptides and identified a peptide (IRIKIRIK; ie, IK8L) with a broad range of bactericidal activity and low cytotoxicity in vitro. Here, employing an animal model, we investigated the antibacterial effects of IK8L in acute infection and demonstrated that peritoneal injection of IK8L to mice down-regulated inflammatory cytokines, alleviated lung injury, and importantly, decreased mortality compared to sham-injected controls. In addition, a math model was used to evaluate in vivo imaging data and predict infection progression in infected live animals. Mechanistically, IK8L can kill Kp by inhibiting biofilm formation and modulating production of inflammatory cytokines through the STAT3/JAK signaling both in vitro and in vivo. Collectively, these findings reveal that IK8L may have potential for preventing or treating Kp infection.
Collapse
Affiliation(s)
- Shirui Tan
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA ; Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
| | - Changpei Gan
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA ; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Rongpeng Li
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA
| | - Yan Ye
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA
| | - Shuang Zhang
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA ; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xu Wu
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, The Nanos, Singapore
| | - Weimin Fan
- Program of Innovative Cancer Therapeutics, First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, People's Republic of China
| | - Min Wu
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA
| |
Collapse
|
24
|
Prechl J, Czirják L. The endothelial deprotection hypothesis for lupus pathogenesis: the dual role of C1q as a mediator of clearance and regulator of endothelial permeability. F1000Res 2015; 4:24. [DOI: 10.12688/f1000research.6075.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/26/2015] [Indexed: 11/20/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous multifactorial systemic autoimmune disease affecting several organs. SLE can start relatively early in life and results in impaired quality of life and shortened life expectancy because of a gradual disease progression leading to cardiovascular, renal and neoplastic disease. The basic mechanisms of the pathogenesis of the disease still remain to be clarified. It is clear that complement proteins play a key and complex role in the development of SLE. Complement component C1q has been known to be a fundamental component of lupus development, but most explanations focus on its role in apoptotic debris removal. Importantly, C1q was recently found to play a key role in the maintenance of vascular endothelial integrity.We suggest that apoptotic products, endothelial cells and extracellular matrix components, which display negatively charged moieties, compete for binding to molecules of the innate humoral immune response, like C1q. Genetic or acquired factors leading to an increased load of apoptotic cell debris and decrease or absence of C1q therefore interfere with the regulation of endothelial permeability and integrity. Furthermore, we suggest that lupus is the net result of an imbalance between the two functions of immune clearance and vascular endothelial integrity maintenance, an imbalance triggered and sustained by autoimmunity, which skews C1q consumption by IgG-mediated complement classical pathway activation on autoantigens. In this triangle of innate clearance, autoimmunity and endothelial integrity, C1q plays a central role.Hence, we interpret the pathogenesis of lupus by identifying three key components, namely innate immune clearance, autoimmunity and endothelial integrity and we establish a link between these components based on the protective role that innate clearance molecules play in endothelial renewal. By including the vasoprotective role of C1q in the interpretation of SLE development we attempt to provide novel explanations for the symptoms, organ damage, diagnostic and therapeutic difficulties of the disease.
Collapse
|
25
|
Prechl J, Czirják L. The endothelial deprotection hypothesis for lupus pathogenesis: the dual role of C1q as a mediator of clearance and regulator of endothelial permeability. F1000Res 2015; 4:24. [PMID: 25901277 PMCID: PMC4392829 DOI: 10.12688/f1000research.6075.2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2015] [Indexed: 12/31/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous multifactorial systemic autoimmune disease affecting several organs. SLE can start relatively early in life and results in impaired quality of life and shortened life expectancy because of a gradual disease progression leading to cardiovascular, renal and neoplastic disease. The basic mechanisms of the pathogenesis of the disease still remain to be clarified. It is clear that complement proteins play a key and complex role in the development of SLE. Complement component C1q has been known to be a fundamental component of lupus development, but most explanations focus on its role in apoptotic debris removal. Importantly, C1q was recently found to play a key role in the maintenance of vascular endothelial integrity. We suggest that apoptotic products, endothelial cells and extracellular matrix components, which display negatively charged moieties, compete for binding to molecules of the innate humoral immune response, like C1q. Genetic or acquired factors leading to an increased load of apoptotic cell debris and decrease or absence of C1q therefore interfere with the regulation of endothelial permeability and integrity. Furthermore, we suggest that lupus is the net result of an imbalance between the two functions of immune clearance and vascular endothelial integrity maintenance, an imbalance triggered and sustained by autoimmunity, which skews C1q consumption by IgG-mediated complement classical pathway activation on autoantigens. In this triangle of innate clearance, autoimmunity and endothelial integrity, C1q plays a central role. Hence, we interpret the pathogenesis of lupus by identifying three key components, namely innate immune clearance, autoimmunity and endothelial integrity and we establish a link between these components based on the protective role that innate clearance molecules play in endothelial renewal. By including the vasoprotective role of C1q in the interpretation of SLE development we attempt to provide novel explanations for the symptoms, organ damage, diagnostic and therapeutic difficulties of the disease.
Collapse
Affiliation(s)
- József Prechl
- Diagnosticum Zrt, Budapest, 1047, Hungary ; MTA-ELTE Immunology Research Group, Budapest, 1117, Hungary
| | - László Czirják
- Department of Rheumatology and Immunology, Clinic Center, University of Pécs, Pécs, 7632, Hungary
| |
Collapse
|
26
|
Jaillon S, Bonavita E, Gentile S, Rubino M, Laface I, Garlanda C, Mantovani A. The long pentraxin PTX3 as a key component of humoral innate immunity and a candidate diagnostic for inflammatory diseases. Int Arch Allergy Immunol 2014; 165:165-78. [PMID: 25531094 DOI: 10.1159/000368778] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The innate immune system is composed of a cellular arm and a humoral arm. Components of the humoral arm include members of the complement cascade and soluble pattern recognition molecules (PRMs). These PRMs recognize pathogen-associated molecular patterns and are functional ancestors of antibodies, playing a role in complement activation, opsonization and agglutination. Pentraxins consist of a set of multimeric soluble proteins and represent the prototypic components of humoral innate immunity. The prototypic long pentraxin PTX3 is highly conserved in evolution and produced by somatic and innate immune cells after proinflammatory stimuli. PTX3 interacts with a set of self, nonself and modified self ligands and exerts essential roles in innate immunity, inflammation control and matrix deposition. In addition, translational studies suggest that PTX3 may be a useful biomarker of human pathologies complementary to C-reactive protein. In this study, we will review the general functions of pentraxins in innate immunity and inflammation, focusing our attention on the prototypic long pentraxin PTX3.
Collapse
|
27
|
Zhang P, Liu X, Cao X. Extracellular pattern recognition molecules in health and diseases. Cell Mol Immunol 2014; 12:255-7. [PMID: 25195513 DOI: 10.1038/cmi.2014.81] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 08/02/2014] [Indexed: 12/30/2022] Open
|
28
|
Lech M, Lorenz G, Kulkarni OP, Grosser MOO, Stigrot N, Darisipudi MN, Günthner R, Wintergerst MWM, Anz D, Susanti HE, Anders HJ. NLRP3 and ASC suppress lupus-like autoimmunity by driving the immunosuppressive effects of TGF-β receptor signalling. Ann Rheum Dis 2014; 74:2224-35. [PMID: 25135254 DOI: 10.1136/annrheumdis-2014-205496] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 07/30/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVES The NLRP3/ASC inflammasome drives host defence and autoinflammatory disorders by activating caspase-1 to trigger the secretion of mature interleukin (IL)-1β/IL-18, but its potential role in autoimmunity is speculative. METHODS We generated and phenotyped Nlrp3-deficient, Asc-deficient, Il-1r-deficient and Il-18-deficient C57BL/6-lpr/lpr mice, the latter being a mild model of spontaneous lupus-like autoimmunity. RESULTS While lack of IL-1R or IL-18 did not affect the C57BL/6-lpr/lpr phenotype, lack of NLRP3 or ASC triggered massive lymphoproliferation, lung T cell infiltrates and severe proliferative lupus nephritis within 6 months, which were all absent in age-matched C57BL/6-lpr/lpr controls. Lack of NLRP3 or ASC increased dendritic cell and macrophage activation, the expression of numerous proinflammatory mediators, lymphocyte necrosis and the expansion of most T cell and B cell subsets. In contrast, plasma cells and autoantibody production were hardly affected. This unexpected immunosuppressive effect of NLRP3 and ASC may relate to their known role in SMAD2/3 phosphorylation during tumour growth factor (TGF)-β receptor signalling, for example, Nlrp3-deficiency and Asc-deficiency significantly suppressed the expression of numerous TGF-β target genes in C57BL/6-lpr/lpr mice and partially recapitulated the known autoimmune phenotype of Tgf-β1-deficient mice. CONCLUSIONS These data identify a novel non-canonical immunoregulatory function of NLRP3 and ASC in autoimmunity.
Collapse
Affiliation(s)
- Maciej Lech
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Georg Lorenz
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Onkar P Kulkarni
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Marian O O Grosser
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Nora Stigrot
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Murthy N Darisipudi
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Roman Günthner
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Maximilian W M Wintergerst
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - David Anz
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Heni Eka Susanti
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| |
Collapse
|
29
|
Lee JK, Park SC, Hahm KS, Park Y. A helix-PXXP-helix peptide with antibacterial activity without cytotoxicity against MDRPA-infected mice. Biomaterials 2013; 35:1025-39. [PMID: 24176194 DOI: 10.1016/j.biomaterials.2013.10.035] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 10/08/2013] [Indexed: 10/26/2022]
Abstract
In response to the growing problem of multidrug-resistant pathogenic microbes, much attention is being paid to naturally occurring and synthetic antimicrobial peptides (AMPs) and the effects of their structural modification. Among these modifications, amino acid substitution is a simple approach to enhancing biological activity and reducing cytotoxicity. An earlier study indicated that HPA3, an analog of HP (2-20) derived from the N-terminus of Helicobacter pylori ribosomal protein L1, forms large pores and shows considerable cytotoxicity. However, HPA3P, in which a proline (Pro) is substituted for glutamic acid (Glu) at position 9 of HPA3, shows markedly less cytotoxicity. This may be attributable to the presence of a Pro-kink into middle of the HPA3P structure within the membrane environment. Unfortunately, HPA3P is not an effective antibacterial agent in vivo. We therefore designed a helix-PXXP-helix structure (HPA3P2), in which Pro was substituted for the Glu and phenylalanine (Phe) at positions 9 and 12 of HPA3, yielding a molecule with a flexible central hinge. As compared to HPA3P, HPA3P3 exhibited dramatically increased antibacterial activity in vivo. ICR mice infected with clinically isolated multidrug-resistant Pseudomonas aeruginosa showed 100% survival when administered one 0.5-mg/kg dose of HPA3P2 or three 0.1-mg/kg doses of HPA3P2. Moreover, in a mouse model of septic shock induced by P. aeruginosa LPS, HPA3P2 reduced production of pro-inflammatory mediators and correspondingly reduced lung (alveolar) and liver tissue damage. The changes in HPA3 behavior with the introduction of Pro likely reflects alterations of the mechanism of action: i) HPA3 forms pores in the bacterial cell membranes, ii) HPA3P permeates the cell membranes and binds to intracellular RNA and DNA, and iii) HPA3P2 acts on the outer cellular membrane component LPS. Collectively, these results suggest HPA3P2 has the potential to be an effective antibiotic for use against multidrug-resistant bacterial strains.
Collapse
Affiliation(s)
- Jong-Kook Lee
- Research Center for Proteinaceous Materials (RCPM), Chosun University, Kwangju 501-759, Republic of Korea
| | | | | | | |
Collapse
|
30
|
Abstract
Lupus nephritis is an immune complex GN that develops as a frequent complication of SLE. The pathogenesis of lupus nephritis involves a variety of pathogenic mechanisms. The extrarenal etiology of systemic lupus is based on multiple combinations of genetic variants that compromise those mechanisms normally assuring immune tolerance to nuclear autoantigens. This loss of tolerance becomes clinically detectable by the presence of antinuclear antibodies. In addition, nucleic acids released from netting or apoptotic neutrophils activate innate and adaptive immunity via viral nucleic acid-specific Toll-like receptors. Therefore, many clinical manifestations of systemic lupus resemble those of viral infection. In lupus, endogenous nuclear particles trigger IFN-α signaling just like viral particles during viral infection. As such, dendritic cells, T helper cells, B cells, and plasma cells all contribute to the aberrant polyclonal autoimmunity. The intrarenal etiology of lupus nephritis involves antibody binding to multiple intrarenal autoantigens rather than the deposition of circulating immune complexes. Tertiary lymphoid tissue formation and local antibody production add to intrarenal complement activation as renal immunopathology progresses. Here we provide an update on the pathogenic mechanisms that lead to lupus nephritis and provide the rationale for the latest and novel treatment strategies.
Collapse
Affiliation(s)
- Maciej Lech
- Department of Nephrology, Medical Clinic and Polyclinic IV, University of Munich, Germany
| | | |
Collapse
|
31
|
PTX3 as a paradigm for the interaction of pentraxins with the complement system. Semin Immunol 2013; 25:79-85. [PMID: 23747040 DOI: 10.1016/j.smim.2013.05.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 05/05/2013] [Indexed: 11/21/2022]
Abstract
Pentraxins are highly conserved components of the humoral arm of innate immunity. They include the short pentraxins C reactive protein (CRP) and serum amyloid P component (SAP), and the long pentraxin PTX3. These are soluble pattern-recognition molecules that are present in the blood and body fluids, and share the ability to recognize pathogens and promote their disposal. CRP and SAP are produced systemically in the liver while PTX3 is produced locally in a number of tissues, macrophages and neutrophils being major sources of this long pentraxin. Pentraxins interact with components of the classical and lectin pathways of Complement as well as with Complement regulators. In particular, PTX3 recognizes C1q, factor H, MBL and ficolins, where these interactions amplify the repertoire of microbial recognition and effector functions of the Complement system. The complex interaction of pentraxins with the Complement system at different levels has broad implications for host defence and regulation of inflammation.
Collapse
|
32
|
Mantovani A, Valentino S, Gentile S, Inforzato A, Bottazzi B, Garlanda C. The long pentraxin PTX3: a paradigm for humoral pattern recognition molecules. Ann N Y Acad Sci 2013; 1285:1-14. [PMID: 23527487 DOI: 10.1111/nyas.12043] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pattern recognition molecules (PRMs) are components of the humoral arm of innate immunity; they recognize pathogen-associated molecular patterns (PAMP) and are functional ancestors of antibodies, promoting complement activation, opsonization, and agglutination. In addition, several PRMs have a regulatory function on inflammation. Pentraxins are a family of evolutionarily conserved PRMs characterized by a cyclic multimeric structure. On the basis of structure, pentraxins have been operationally divided into short and long families. C-reactive protein (CRP) and serum amyloid P component are prototypes of the short pentraxin family, while pentraxin 3 (PTX3) is a prototype of the long pentraxins. PTX3 is produced by somatic and immune cells in response to proinflammatory stimuli and Toll-like receptor engagement, and it interacts with several ligands and exerts multifunctional properties. Unlike CRP, PTX3 gene organization and regulation have been conserved in evolution, thus allowing its pathophysiological roles to be evaluated in genetically modified animals. Here we will briefly review the general properties of CRP and PTX3 as prototypes of short and long pentraxins, respectively, emphasizing in particular the functional role of PTX3 as a prototypic PRM with antibody-like properties.
Collapse
|
33
|
Biology of Human Pentraxin 3 (PTX3) in Acute and Chronic Kidney Disease. J Clin Immunol 2013; 33:881-90. [DOI: 10.1007/s10875-013-9879-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 02/18/2013] [Indexed: 12/13/2022]
|
34
|
Sjöwall C, Olin AI, Skogh T, Wetterö J, Mörgelin M, Nived O, Sturfelt G, Bengtsson AA. C-reactive protein, immunoglobulin G and complement co-localize in renal immune deposits of proliferative lupus nephritis. Autoimmunity 2013; 46:205-14. [DOI: 10.3109/08916934.2013.764992] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
35
|
Endogenous and exogenous pentraxin-3 limits postischemic acute and chronic kidney injury. Kidney Int 2013; 83:647-61. [PMID: 23325083 DOI: 10.1038/ki.2012.463] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Ischemia-reperfusion activates innate immunity and sterile inflammation, resulting in acute kidney injury. Since pentraxin 3 (PTX3) regulates multiple aspects of innate immunity and tissue inflammation, we tested whether PTX3 would be involved in renal ischemia-reperfusion injury. Renal pedicle clamping increased PTX3 serum levels, as well as PTX3 expression, inside the kidney but predominantly in CD45/CD11c(+) cells, a subpopulation of intrarenal mononuclear phagocytes. Lack of PTX3 aggravated postischemic acute kidney injury as evidenced by massive tubular necrosis, and TNF and IL-6 release, as well as massively increased neutrophil and macrophage infiltrates at 24 h. This was followed by tubular atrophy, interstitial fibrosis, and kidney shrinking 10 weeks later. In vivo microscopy uncovered increased leukocyte adhesion and transmigration in postischemic microvessels of Ptx3-deficient mice. Furthermore, injection of recombinant PTX3 up to 6 h after reperfusion prevented renal leukocyte recruitment and postischemic kidney injury. Thus, local PTX3 release from a subpopulation of intrarenal mononuclear phagocytes or delayed PTX3 treatment limits postischemic renal inflammation. Conversely, Ptx3 loss-of-function mutations predispose to postischemic acute kidney injury and subsequent chronic kidney disease.
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Lupus nephritis is a complex autoimmune disease that develops its own dynamic upon damaging the renal ultrastructure. Here, we summarize the latest pathophysiological concepts of lupus nephritis and how these translate into novel therapeutic options. RECENT FINDINGS Multidisciplinary research activities form a better understanding about how lupus develops from an unfortunate combination of gene variants that promote the loss of tolerance, that impair the clearance of dying cells, that regulate the immune interpretation of autoantigens as well as the peripheral control of autoreactive lymphocytes. As a new entry, nuclear particles also act as autoadjuvants mimicking viral particles and triggering interferon-alpha-dependent antiviral immune responses that cause symptoms similar to viral infection. SUMMARY A set of novel drugs have the potential to more specifically interfere with these pathomechanisms and raise hope to more efficiently treat lupus nephritis with fewer side-effects in the near future.
Collapse
|
37
|
Inforzato A, Reading PC, Barbati E, Bottazzi B, Garlanda C, Mantovani A. The "sweet" side of a long pentraxin: how glycosylation affects PTX3 functions in innate immunity and inflammation. Front Immunol 2013; 3:407. [PMID: 23316195 PMCID: PMC3539679 DOI: 10.3389/fimmu.2012.00407] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 12/17/2012] [Indexed: 12/24/2022] Open
Abstract
Innate immunity represents the first line of defense against pathogens and plays key roles in activation and orientation of the adaptive immune response. The innate immune system comprises both a cellular and a humoral arm. Components of the humoral arm include soluble pattern recognition molecules (PRMs) that recognize pathogen-associated molecular patterns and initiate the immune response in coordination with the cellular arm, therefore acting as functional ancestors of antibodies. The long pentraxin PTX3 is a prototypic soluble PRM that is produced at sites of infection and inflammation by both somatic and immune cells. Gene targeting of this evolutionarily conserved protein has revealed a non-redundant role in resistance to selected pathogens. Moreover, PTX3 exerts important functions at the crossroad between innate immunity, inflammation, and female fertility. The human PTX3 protein contains a single N-glycosylation site that is fully occupied by complex type oligosaccharides, mainly fucosylated and sialylated biantennary glycans. Glycosylation has been implicated in a number of PTX3 activities, including neutralization of influenza viruses, modulation of the complement system, and attenuation of leukocyte recruitment. Therefore, this post translational modification might act as a fine tuner of PTX3 functions in native immunity and inflammation. Here we review the studies on PTX3, with emphasis on the glycan-dependent mechanisms underlying pathogen recognition and crosstalk with other components of the innate immune system.
Collapse
Affiliation(s)
- Antonio Inforzato
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center Rozzano, Italy
| | | | | | | | | | | |
Collapse
|
38
|
Lech M, Rommele C, Anders HJ. Pentraxins in nephrology: C-reactive protein, serum amyloid P and pentraxin-3. Nephrol Dial Transplant 2012; 28:803-11. [PMID: 23243042 DOI: 10.1093/ndt/gfs448] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Every clinician uses C-reactive protein (CRP) levels as a biomarker for systemic inflammation in acute disorders. Nephrologists also consider CRP levels as a predictor for overall mortality in patients with chronic kidney disease or end-stage renal disease. But what is the biological function of CRP? CRP is a member of the family of pentraxins, which are small pentameric innate immunity effector proteins. Pentraxins are absent or weakly expressed during homeostasis. However, the pro-inflammatory cytokines interleukin (IL)-1, IL-6 and tumour necrosis factor induce CRP and serum amyloid P (SAP) in hepatocytes, whereas the long pentraxins, such as pentraxin (PTX)-3, are produced in peripheral tissues and monocytic phagocytes. Pentraxins opsonize pathogens or other particles such as dead cells, for their phagocytic clearance or induce pathogen killing in extracellular compartments. In this review, we discuss the immunoregulatory properties of the different members of the pentraxin family. We discuss the evolving evidence demonstrating their roles in acute and chronic forms of kidney disease and the significance of SAP and PTX3 as additional biomarkers of innate immune activation and systemic inflammation.
Collapse
Affiliation(s)
- Maciej Lech
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, University of Munich-LMU, Ziemssenstr 1 D-80336, Munich, Germany
| | | | | |
Collapse
|
39
|
Daigo K, Hamakubo T. Host-protective effect of circulating pentraxin 3 (PTX3) and complex formation with neutrophil extracellular traps. Front Immunol 2012; 3:378. [PMID: 23248627 PMCID: PMC3521240 DOI: 10.3389/fimmu.2012.00378] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 11/26/2012] [Indexed: 01/21/2023] Open
Abstract
Pentraxin 3 (PTX3) is a soluble pattern recognition receptor which is classified as a long-pentraxin in the pentraxin family. It is known to play an important role in innate immunity, inflammatory regulation, and female fertility. PTX3 is synthesized by specific cells, primarily in response to inflammatory signals. Among these various cells, neutrophils have a unique PTX3 production system. Neutrophils store PTX3 in neutrophil-specific granules and then the stored PTX3 is released and localizes in neutrophil extracellular traps (NETs). Although certain NET components have been identified, such as histones and anti-microbial proteins, the detailed mechanisms by which NETs localize, as well as capture and kill microbes, have not been fully elucidated. PTX3 is a candidate diagnostic marker of infection and vascular damage. In severe infectious diseases such as sepsis, the circulating PTX3 concentration increases greatly (up to 100 ng/mL, i.e., up to 100-fold of the normal level). Even though it is clearly implied that PTX3 plays a protective role in sepsis and certain other disorders, the detailed mechanisms by which it does so remain unclear. A proteomic study of PTX3 ligands in septic patients revealed that PTX3 forms a complex with certain NET component proteins. This suggests a role for PTX3 in which it facilitates the efficiency of anti-microbial protein pathogen clearance by interacting with both pathogens and anti-microbial proteins. We discuss the possible relationships between PTX3 and NET component proteins in the host protection afforded by the innate immune response. The PTX3 complex has the potential to be a highly useful diagnostic marker of sepsis and other inflammatory diseases.
Collapse
Affiliation(s)
- Kenji Daigo
- Department of Molecular Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo Tokyo, Japan
| | | |
Collapse
|
40
|
Tissues use resident dendritic cells and macrophages to maintain homeostasis and to regain homeostasis upon tissue injury: the immunoregulatory role of changing tissue environments. Mediators Inflamm 2012; 2012:951390. [PMID: 23251037 PMCID: PMC3518145 DOI: 10.1155/2012/951390] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 10/25/2012] [Indexed: 01/07/2023] Open
Abstract
Most tissues harbor resident mononuclear phagocytes, that is, dendritic cells and macrophages. A classification that sufficiently covers their phenotypic heterogeneity and plasticity during homeostasis and disease does not yet exist because cell culture-based phenotypes often do not match those found in vivo. The plasticity of mononuclear phagocytes becomes obvious during dynamic or complex disease processes. Different data interpretation also originates from different conceptual perspectives. An immune-centric view assumes that a particular priming of phagocytes then causes a particular type of pathology in target tissues, conceptually similar to antigen-specific T-cell priming. A tissue-centric view assumes that changing tissue microenvironments shape the phenotypes of their resident and infiltrating mononuclear phagocytes to fulfill the tissue's need to maintain or regain homeostasis. Here we discuss the latter concept, for example, why different organs host different types of mononuclear phagocytes during homeostasis. We further discuss how injuries alter tissue environments and how this primes mononuclear phagocytes to enforce this particular environment, for example, to support host defense and pathogen clearance, to support the resolution of inflammation, to support epithelial and mesenchymal healing, and to support the resolution of fibrosis to the smallest possible scar. Thus, organ- and disease phase-specific microenvironments determine macrophage and dendritic cell heterogeneity in a temporal and spatial manner, which assures their support to maintain and regain homeostasis in whatever condition. Mononuclear phagocytes contributions to tissue pathologies relate to their central roles in orchestrating all stages of host defense and wound healing, which often become maladaptive processes, especially in sterile and/or diffuse tissue injuries.
Collapse
|
41
|
Doni A, Garlanda C, Bottazzi B, Meri S, Garred P, Mantovani A. Interactions of the humoral pattern recognition molecule PTX3 with the complement system. Immunobiology 2012; 217:1122-8. [PMID: 22964239 DOI: 10.1016/j.imbio.2012.07.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 07/16/2012] [Indexed: 12/20/2022]
Abstract
The innate immune system comprises a cellular and a humoral arm. The long pentraxin PTX3 is a fluid phase pattern recognition molecule, which acts as an essential component of the humoral arm of innate immunity. PTX3 has antibody-like properties including interactions with complement components. PTX3 interacts with C1q, ficolin-1 and ficolin-2 as well as mannose-binding lectin, recognition molecules in the classical and lectin complement pathways. The formation of these heterocomplexes results in cooperative pathogen recognition and complement activation. Interactions with C4b binding protein and factor H, the principal regulators of the classical, lectin and alternative complement pathways, show that PTX3 also may have a major influence on the regulation of the complement system. The complex interaction of PTX3 with the complement system at different levels has broad implications for host defence and regulation of inflammation.
Collapse
Affiliation(s)
- Andrea Doni
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Innate immunity represents the first line of defence against pathogens and plays key roles in activation and orientation of the adaptive immune response. The innate immune system comprises both a cellular and a humoral arm. Components of the humoral arm include soluble pattern recognition molecules (PRMs) that recognise pathogens associated molecular patterns (PAMPs) and initiate the immune response in coordination with the cellular arm, therefore acting as functional ancestors of antibodies. The long pentraxin PTX3 is a prototypic soluble PRM that is produced at sites of infection and inflammation by both somatic and immune cells. Gene targeting of this evolutionarily conserved protein has revealed a non-redundant role in resistance to selected pathogens. Moreover, PTX3 exerts important functions at the crossroad between innate immunity, inflammation and female fertility. Here we review the studies on PTX3, with emphasis on pathogen recognition and crosstalk with other components of the innate immune system.
Collapse
|