1
|
Dellière S, Chauvin C, Wong SSW, Gressler M, Possetti V, Parente R, Fontaine T, Krüger T, Kniemeyer O, Bayry J, Carvalho A, Brakhage AA, Inforzato A, Latgé JP, Aimanianda V. Interplay between host humoral pattern recognition molecules controls undue immune responses against Aspergillus fumigatus. Nat Commun 2024; 15:6966. [PMID: 39138196 PMCID: PMC11322389 DOI: 10.1038/s41467-024-51047-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
Pentraxin 3 (PTX3), a long pentraxin and a humoral pattern recognition molecule (PRM), has been demonstrated to be protective against Aspergillus fumigatus, an airborne human fungal pathogen. We explored its mode of interaction with A. fumigatus, and the resulting implications in the host immune response. Here, we demonstrate that PTX3 interacts with A. fumigatus in a morphotype-dependent manner: (a) it recognizes germinating conidia through galactosaminogalactan, a surface exposed cell wall polysaccharide of A. fumigatus, (b) in dormant conidia, surface proteins serve as weak PTX3 ligands, and (c) surfactant protein D (SP-D) and the complement proteins C1q and C3b, the other humoral PRMs, enhance the interaction of PTX3 with dormant conidia. SP-D, C3b or C1q opsonized conidia stimulated human primary immune cells to release pro-inflammatory cytokines and chemokines. However, subsequent binding of PTX3 to SP-D, C1q or C3b opsonized conidia significantly decreased the production of pro-inflammatory cytokines/chemokines. PTX3 opsonized germinating conidia also significantly lowered the production of pro-inflammatory cytokines/chemokines while increasing IL-10 (an anti-inflammatory cytokine) released by immune cells when compared to the unopsonized counterpart. Overall, our study demonstrates that PTX3 recognizes A. fumigatus either directly or by interplaying with other humoral PRMs, thereby restraining detrimental inflammation. Moreover, PTX3 levels were significantly higher in the serum of patients with invasive pulmonary aspergillosis (IPA) and COVID-19-associated pulmonary aspergillosis (CAPA), supporting previous observations in IPA patients, and suggesting that it could be a potential panel-biomarker for these pathological conditions caused by A. fumigatus.
Collapse
Affiliation(s)
- Sarah Dellière
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Unité Mycologie Moléculaire, Paris, France
- Laboratoire de Parasitologie-Mycologie, AP-HP, Hôpital Saint-Louis, Paris, France
- Institut Pasteur, Université Paris Cité, Immunobiology of Aspergillus, Paris, France
| | - Camille Chauvin
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris-Cité, Paris, France
| | - Sarah Sze Wah Wong
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Unité Mycologie Moléculaire, Paris, France
- Institut Pasteur, Unité des Aspergillus, Paris, France
| | - Markus Gressler
- Institut Pasteur, Unité des Aspergillus, Paris, France
- Faculty of Biological Sciences, Pharmaceutical Microbiology, Friedrich Schiller University Jena, Winzerlaer Strasse 2, 07745, Jena, Germany; Pharmaceutical Microbiology, Leibniz Institute for Natural Product Research, and Infection Biology-Hans-Knöll-Institute, Winzerlaer Strasse 2, Jena, Germany
| | - Valentina Possetti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Thierry Fontaine
- Institut Pasteur, Unité des Aspergillus, Paris, France
- Institut Pasteur, Université Paris Cité, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research, and Infection Biology (Leibniz-HKI), Jena, Germany
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research, and Infection Biology (Leibniz-HKI), Jena, Germany
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris-Cité, Paris, France
- Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Palakkad, India
| | - Agostinho Carvalho
- Life & Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Axel A Brakhage
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research, and Infection Biology (Leibniz-HKI), Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Antonio Inforzato
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Vishukumar Aimanianda
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Unité Mycologie Moléculaire, Paris, France.
- Institut Pasteur, Université Paris Cité, Immunobiology of Aspergillus, Paris, France.
- Institut Pasteur, Unité des Aspergillus, Paris, France.
| |
Collapse
|
2
|
Santos-Ribeiro D, Cunha C, Carvalho A. Humoral pathways of innate immune regulation in granuloma formation. Trends Immunol 2024; 45:419-427. [PMID: 38762333 DOI: 10.1016/j.it.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/20/2024]
Abstract
The humoral arm of mammalian innate immunity regulates several molecular mechanisms involved in resistance to pathogens, inflammation, and tissue repair. Recent studies highlight the crucial role played by humoral mediators in granulomatous inflammation. However the molecular mechanisms linking the function of these soluble molecules to the initiation and maintenance of granulomas remain elusive. We propose that humoral innate immunity coordinates fundamental physiological processes in macrophages which, in turn, initiate activation and transformation events that enable granuloma formation. We discuss the involvement of humoral mediators in processes such as immune activation, phagocytosis, metabolism, and tissue remodeling, and how these can dictate macrophage functionality during granuloma formation. These advances present opportunities for discovering novel disease factors and developing targeted, more effective treatments for granulomatous diseases.
Collapse
Affiliation(s)
- Diana Santos-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
3
|
Giacomini A, Turati M, Grillo E, Rezzola S, Ghedini GC, Schuind AC, Foglio E, Maccarinelli F, Faletti J, Filiberti S, Chambery A, Valletta M, Melocchi L, Gofflot S, Chiavarina B, Turtoi A, Presta M, Ronca R. The PTX3/TLR4 autocrine loop as a novel therapeutic target in triple negative breast cancer. Exp Hematol Oncol 2023; 12:82. [PMID: 37749607 PMCID: PMC10519006 DOI: 10.1186/s40164-023-00441-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/04/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND The pattern recognition receptor long pentraxin-3 (PTX3) plays conflicting roles in cancer by acting as an oncosuppressor or as a pro-tumor mediator depending on tumor context. Triple negative breast cancer (TNBC) represents the most aggressive histotype of breast cancer, characterized by the lack of efficacious therapeutic targets/approaches and poor prognosis. Thus, the characterization of new molecular pathways and/or alternative druggable targets is of great interest in TNBC. METHODS The expression of PTX3 in BC tumor samples and in BC cell lines has been analyzed using the Gene Expression-Based Outcome for Breast Cancer Online (GOBO), qPCR, Western blot and ELISA assay. The contribution of tumor and stromal cells to PTX3 production in TNBC was assessed by analyzing single cell RNA sequencing data and RNAscope performed on TNBC tumor samples. In order to investigate the effects of PTX3 in TNBC, different cell lines were engineered to knock-down (MDA-MB-231 and BT549 cells) or overexpress (MDA-MB-468 and E0771 cells) PTX3. Finally, using these engineered cells, in vitro (including gene expression profiling and gene set enrichment analyses) and in vivo (orthotopic tumor models in immune-compromised and immune competent mice) analyses were performed to assess the role and the molecular mechanism(s) exerted by PTX3 in TNBC. RESULTS In silico and experimental data indicate that PTX3 is mainly produced by tumor cells in TNBC and that its expression levels correlate with tumor stage. Accordingly, gene expression and in vitro results demonstrate that PTX3 overexpression confers a high aggressive/proliferative phenotype and fosters stem-like features in TNBC cells. Also, PTX3 expression induces a more tumorigenic potential when TNBC cells are grafted orthotopically in vivo. Conversely, PTX3 downregulation results in a less aggressive behavior of TNBC cells. Mechanistically, our data reveal that PTX3 drives the activation of the pro-tumorigenic Toll-like receptor 4 (TLR4) signaling pathway in TNBC, demonstrating for the first time that the PTX3/TLR4 autocrine stimulation loop contributes to TNBC aggressiveness and that TLR4 inhibition significantly impacts the growth of PTX3-producing TNBC cells. CONCLUSION Altogether, these data shed light on the role of tumor-produced PTX3 in TNBC and uncover the importance of the PTX3/TLR4 axis for therapeutic and prognostic exploitation in TNBC.
Collapse
Affiliation(s)
- Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Marta Turati
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia Cristina Ghedini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Ander Churruca Schuind
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Eleonora Foglio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Federica Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Jessica Faletti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Serena Filiberti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Angela Chambery
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), University of Campania 'Luigi Vanvitelli', Caserta, Italy
| | - Mariangela Valletta
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), University of Campania 'Luigi Vanvitelli', Caserta, Italy
| | - Laura Melocchi
- Pathology Unit, Fondazione Poliambulanza Hospital Institute, Brescia, 25121, Italy
| | | | - Barbara Chiavarina
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, University of Montpellier, Montpellier, France
| | - Andrei Turtoi
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, University of Montpellier, Montpellier, France
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
4
|
Gupta SK, Wilson T, Maclean PH, Rehm BHA, Heiser A, Buddle BM, Wedlock DN. Mycobacterium avium subsp. paratuberculosis antigens induce cellular immune responses in cattle without causing reactivity to tuberculin in the tuberculosis skin test. Front Immunol 2023; 13:1087015. [PMID: 36741398 PMCID: PMC9889921 DOI: 10.3389/fimmu.2022.1087015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/27/2022] [Indexed: 01/19/2023] Open
Abstract
Mycobacterium avium subspecies paratuberculosis (MAP) causes chronic progressive granulomatous enteritis leading to diarrhea, weight-loss, and eventual death in ruminants. Commercially available vaccine provides only partial protection against MAP infection and can interfere with the use of current diagnostic tests for bovine tuberculosis in cattle. Here, we characterized immune responses in calves to vaccines containing four truncated MAP antigens as a fusion (Ag85A202-347-SOD1-72-Ag85B173-330-74F1-148+669-786), either displayed on protein particles, or expressed as a soluble recombinant MAP (rMAP) fusion protein as well as to commercially available Silirum® vaccine. The rMAP fusion protein elicited the strongest antigen-specific antibody responses to both PPDA and recombinant antigen and strong and long-lasting T-cell immune responses to these antigens, as indicated by increased production of IFN-γ and IL-17A in antigen-stimulated whole blood cultures. The MAP fusion protein particle vaccine induced minimal antibody responses and weak IFN-γ responses but stimulated IL-17A responses to recombinant antigen. The immune response profile of Silirum® vaccine was characterized by weak antibodies and strong IFN-γ and IL-17A responses to PPDA. Transcription analysis on antigen-stimulated leukocytes from cattle vaccinated with rMAP fusion protein showed differential expression of several immune response genes and genes involved in costimulatory signaling, TLR4, TLR2, PTX3, PTGS2, PD-L1, IL1B, IL2, IL6, IL12B, IL17A, IL22, IFNG, CD40, and CD86. Moreover, the expression of several genes of immune pathways correlated with cellular immune responses in the rMAP fusion protein vaccinated group. These genes have key roles in pathways of mycobacterial immunity, including autophagy, manipulation of macrophage-mediated killing, Th17- and regulatory T cells- (Treg) mediated responses. Calves vaccinated with either the rMAP fusion protein or MAP fusion protein particle vaccine did not induce reactivity to PPDA and PPDB in a comparative cervical skin test, whereas Silirum® induced reactivity to these tuberculins in most of the vaccinated animals. Overall, our results suggest that a combination of recombinant MAP antigens in the form of a soluble fusion protein vaccine are capable of inducing strong antigen-specific humoral and a balanced Th1/Th17-cell immune response. These findings, together with the absence of reactivity to tuberculin, suggest this subunit vaccine could provide protective immunity against intracellular MAP infection in cattle without compromising the use of current bovine tuberculosis surveillance test.
Collapse
Affiliation(s)
- Sandeep K. Gupta
- AgResearch, Hopkirk Research Institute, Palmerston North, New Zealand,*Correspondence: Sandeep K. Gupta,
| | - Tania Wilson
- AgResearch, Hopkirk Research Institute, Palmerston North, New Zealand
| | | | - Bernd H. A. Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia,Menzies Health Institute Queensland (MHIQ), Griffith University, Gold Coast, QLD, Australia
| | - Axel Heiser
- AgResearch, Hopkirk Research Institute, Palmerston North, New Zealand
| | - Bryce M. Buddle
- AgResearch, Hopkirk Research Institute, Palmerston North, New Zealand
| | - D. Neil Wedlock
- AgResearch, Hopkirk Research Institute, Palmerston North, New Zealand
| |
Collapse
|
5
|
Molecular insight into pentraxin-3: update advances in innate immunity, inflammation, tissue remodeling, diseases, and drug role. Biomed Pharmacother 2022; 156:113783. [DOI: 10.1016/j.biopha.2022.113783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
|
6
|
Soltani N, Esmaeil N, Marandi S, Hovsepian V, Momen T, Shahsanai A. A 2-week combined high-intensity interval training regulates inflammatory status in young females with obesity. Sci Sports 2022. [DOI: 10.1016/j.scispo.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
7
|
Aspergillus Endophthalmitis: Epidemiology, Pathobiology, and Current Treatments. J Fungi (Basel) 2022; 8:jof8070656. [PMID: 35887412 PMCID: PMC9318612 DOI: 10.3390/jof8070656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/21/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Abstract
Fungal endophthalmitis is one of the leading causes of vision loss worldwide. Post-operative and traumatic injuries are major contributing factors resulting in ocular fungal infections in healthy and, more importantly, immunocompromised individuals. Among the fungal pathogens, the Aspergillus species, Aspergillus fumigatus, continues to be more prevalent in fungal endophthalmitis patients. However, due to overlapping clinical symptoms with other endophthalmitis etiology, fungal endophthalmitis pose a challenge in its diagnosis and treatment. Hence, it is critical to understand its pathobiology to develop and deploy proper therapeutic interventions for combating Aspergillus infections. This review highlights the different modes of Aspergillus transmission and the host immune response during endophthalmitis. Additionally, we discuss recent advancements in the diagnosis of fungal endophthalmitis. Finally, we comprehensively summarize various antifungal regimens and surgical options for the treatment of Aspergillus endophthalmitis.
Collapse
|
8
|
Gonçalves SM, Cunha C, Carvalho A. Understanding the genetic basis of immune responses to fungal infection. Expert Rev Anti Infect Ther 2022; 20:987-996. [PMID: 35385368 DOI: 10.1080/14787210.2022.2063839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Fungal infections represent a global public health problem that affect millions of people. Despite remarkable advances achieved over the last decades, available diagnostic and therapeutic tools remain insufficient for the optimal management of these diseases. The clinical course of fungal infection is highly variable, and evidence accumulated from patients with rare mutations and cohort-based studies suggests that the trajectory of disease is largely defined by patient genetics and its impact on immune responses. Therefore, there is an urgent need to elucidate the precise mechanisms by which which genetic variants influence the risk, progression, and outcome of fungal infection. AREAS COVERED In this review, we highlight recent advances in our understanding of the genetic factors that influence antifungal immune responses based on candidate gene studies and genome-wide approaches performed in different experimental and clinical models. EXPERT OPINION Research on genetics of susceptibility to infection is expected to lead to a detailed knowledge framework for the pathogenesis of human fungal infections and unveil novel targets and pathways amenable to clinical intervention.
Collapse
Affiliation(s)
- Samuel M Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
9
|
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
|
10
|
Bigot J, Guillot L, Guitard J, Ruffin M, Corvol H, Balloy V, Hennequin C. Bronchial Epithelial Cells on the Front Line to Fight Lung Infection-Causing Aspergillus fumigatus. Front Immunol 2020; 11:1041. [PMID: 32528481 PMCID: PMC7257779 DOI: 10.3389/fimmu.2020.01041] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
Aspergillus fumigatus is an environmental filamentous fungus that can be pathogenic for humans, wherein it is responsible for a large variety of clinical forms ranging from allergic diseases to life-threatening disseminated infections. The contamination occurs by inhalation of conidia present in the air, and the first encounter of this fungus in the human host is most likely with the bronchial epithelial cells. Although alveolar macrophages have been widely studied in the Aspergillus–lung interaction, increasing evidence suggests that bronchial epithelium plays a key role in responding to the fungus. This review focuses on the innate immune response of the bronchial epithelial cells against A. fumigatus, the predominant pathogenic species. We have also detailed the molecular interactants and the effects of the different modes of interaction between these cells and the fungus.
Collapse
Affiliation(s)
- Jeanne Bigot
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Parasitologie-Mycologie, Paris, France
| | - Loïc Guillot
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| | - Juliette Guitard
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Parasitologie-Mycologie, Paris, France
| | - Manon Ruffin
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| | - Harriet Corvol
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Trousseau, Service de Pneumologie Pédiatrique, Paris, France
| | - Viviane Balloy
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| | - Christophe Hennequin
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Parasitologie-Mycologie, Paris, France
| |
Collapse
|
11
|
Borghi M, Puccetti M, Pariano M, Renga G, Stincardini C, Ricci M, Giovagnoli S, Costantini C, Romani L. Tryptophan as a Central Hub for Host/Microbial Symbiosis. Int J Tryptophan Res 2020; 13:1178646920919755. [PMID: 32435131 PMCID: PMC7225782 DOI: 10.1177/1178646920919755] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 03/14/2020] [Indexed: 01/06/2023] Open
Abstract
Amino acid catabolism occurs during inflammation and regulates innate and adaptive immunity. The role of commensal bacteria in amino acid catabolism and the production of metabolites able to regulate the development and function of the innate immune system is increasingly being recognized. Therefore, commensal bacteria are key players in the maintenance of immune homeostasis. However, the intestinal microbiota also contributes to susceptibility and response to infectious diseases. This is self-evident for fungal infections known to occur as a consequence of weakened immune system and broad-spectrum antibiotic use or abuse. Thus, diseases caused by opportunistic fungi can no longer be viewed as dependent only on a weakened host but also on a disrupted microbiota. Based on these premises, the present review focuses on the role of amino acid metabolic pathways in the dialogue between the mammalian host and its microbiota and the potential implications in fungal commensalism and infectivity.
Collapse
Affiliation(s)
- Monica Borghi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Science, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Maurizio Ricci
- Department of Pharmaceutical Science, University of Perugia, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Science, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
12
|
Magrone T, Jirillo E, Magrone M, Russo MA, Romita P, Massari F, Foti C. Red Grape Polyphenol Oral Administration Improves Immune Response in Women Affected by Nickel-Mediated Allergic Contact Dermatitis. Endocr Metab Immune Disord Drug Targets 2020; 21:374-384. [PMID: 32167433 DOI: 10.2174/1871530320666200313152648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/10/2020] [Accepted: 01/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Our previous findings demonstrated that in vitro supplementation of polyphenols, extracted from seeds of red grape (Nero di Troia cultivar), to peripheral lymphomonocytes from patients affected by allergic contact dermatitis (ACD) to nickel (Ni) could reduce the release of proinflammatory cytokines and nitric oxide (NO), while increasing the levels of interleukin (IL)-10, an anti-inflammatory cytokine. OBJECTIVE To assess whether an intervention with oral administration of polyphenols leads to a reduction of peripheral biomarkers in ACD patients. METHODS At T0, 25 patients affected by ACD to Ni were orally administered with 300 mg polyphenols prodie extracted from seeds of red grape (Nero di Troia cultivar) (NATUR-OX®) for 3 months (T1). The other 25 patients affected by ACD to Ni received placebo only for the same period of time. Serum biomarkers were analyzed at T0 and T1. In both groups, seven dropouts were recorded. RESULTS At T1 in comparison to T0, in treated patients, values of interferon-γ, IL-4, IL-17, pentraxin 3 and NO decreased, while IL-10 levels increased when compared with T0 values. Conversely, in placebo- treated patients, no modifications of biomarkers were evaluated at T1. CONCLUSION Present laboratory data rely on the anti-oxidant, anti-inflammatory and anti-allergic properties of polyphenols.
Collapse
Affiliation(s)
- Thea Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari, Bari, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari, Bari, Italy
| | - Manrico Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari, Bari, Italy
| | - Matteo A Russo
- MEBIC Consortium, San Raffaele Open University of Rome and IRCCS San Raffaele Pisana of Rome, Rome, Italy
| | - Paolo Romita
- Department of Biomedical Sciences and Human Oncology, University of Bari, School of Medicine, University of Bari, Bari, Italy
| | - Francesco Massari
- Department of Biomedical Sciences and Human Oncology, University of Bari, School of Medicine, University of Bari, Bari, Italy
| | - Caterina Foti
- Department of Biomedical Sciences and Human Oncology, University of Bari, School of Medicine, University of Bari, Bari, Italy
| |
Collapse
|
13
|
Parente R, Doni A, Bottazzi B, Garlanda C, Inforzato A. The complement system in Aspergillus fumigatus infections and its crosstalk with pentraxins. FEBS Lett 2020; 594:2480-2501. [PMID: 31994174 DOI: 10.1002/1873-3468.13744] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/19/2019] [Accepted: 01/16/2020] [Indexed: 12/13/2022]
Abstract
Aspergillosis is a life-threatening infection mostly affecting immunocompromised individuals and primarily caused by the saprophytic fungus Aspergillus fumigatus. At the host-pathogen interface, both cellular and humoral components of the innate immune system are increasingly acknowledged as essential players in the recognition and disposal of this opportunistic mold. Fundamental hereof is the contribution of the complement system, which deploys all three activation pathways in the battle against A. fumigatus, and functionally cooperates with other soluble pattern recognition molecules, including pentraxins. In particular, preclinical and clinical observations point to the long pentraxin PTX3 as a nonredundant and complement-dependent effector with protective functions against A. fumigatus. Based on past and current literature, here we discuss how the complement participates in the immune response to this fungal pathogen, and illustrate its crosstalk with the pentraxins, with a focus on PTX3. Emphasis is placed on the molecular mechanisms underlying such processes, the genetic evidence from human epidemiology, and the translational potential of the currently available knowledge.
Collapse
Affiliation(s)
- Raffaella Parente
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute - IRCCS, Milan, Italy
| | - Andrea Doni
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute - IRCCS, Milan, Italy
| | - Barbara Bottazzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute - IRCCS, Milan, Italy
| | - Cecilia Garlanda
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute - IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Antonio Inforzato
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute - IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
14
|
Aerobic Training Down-Regulates Pentraxin 3 and Pentraxin 3/Toll-Like Receptor 4 Ratio, Irrespective of Oxidative Stress Response, in Elderly Subjects. Antioxidants (Basel) 2020; 9:antiox9020110. [PMID: 32012711 PMCID: PMC7070734 DOI: 10.3390/antiox9020110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 01/14/2023] Open
Abstract
Reactive oxygen and nitrogen species-mediated cellular aging has been linked to diseases such as atherothrombosis and cancer. Although pentraxin 3 (PTX3) is associated with aging-related diseases via TLR4-dependent anti-inflammatory effects, its relationship with oxidative stress in aging remains to be elucidated. Exercise is proposed as the key intervention for health maintenance in the elderly. This study aimed to examine the association of PTX3 levels with changes in oxidative stress in both plasma and peripheral blood mononuclear cells (PBMCs), following aerobic training in elderly adults. Nine trained and five controls participated in an eight-week aerobic training protocol. Enzyme-linked immunosorbent assay (ELISA) and Western blot analyses were used to determine PTX3, toll-like receptor 4 (TLR4), and levels of oxidative stress biomarkers [3-nitrotyrosine (3NT), 4-hydroxynonenal (4-HNE), glutathione (GSH), protein carbonyl (PC), reactive oxygen/ nitrogen species (ROS/RNS), and trolox equivalent antioxidant capacity (TEAC)] in plasma and/or PBMCs. Results showed a down-regulation of PTX3 expression in PBMCs following aerobic training, along with decreased PTX3/TLR4 ratios. Oxidative stress responses in PBMCs remained unchanged with the exercise protocol. Comparable levels of plasma PTX3 and oxidative stress biomarkers were observed in trained vs. control groups. No correlation was found between PTX3 and any oxidative stress biomarkers following training. These findings demonstrated the down-regulation of PTX3 and PTX3/TLR4 ratio, irrespective of oxidative stress response, in elderly adults following eight weeks of aerobic training.
Collapse
|
15
|
He Q, Li H, Rui Y, Liu L, He B, Shi Y, Su X. Pentraxin 3 Gene Polymorphisms and Pulmonary Aspergillosis in Chronic Obstructive Pulmonary Disease Patients. Clin Infect Dis 2019; 66:261-267. [PMID: 29020397 DOI: 10.1093/cid/cix749] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/17/2017] [Indexed: 12/20/2022] Open
Abstract
Background Pentraxin 3 (PTX3) plays an important role in resistance to Aspergillus infections. Previous studies have suggested that PTX3 polymorphisms influence the risk of invasive aspergillosis among transplantation recipients. This study investigated the association between PTX3 gene polymorphisms and pulmonary aspergillosis in a chronic obstructive pulmonary disease (COPD) population. Methods We included 173 consecutive inpatients with COPD. Thirty-six case patients were finally diagnosed with pulmonary aspergillosis. Among these, 25 case patients had invasive pulmonary aspergillosis (IPA), and 11 case patients had chronic pulmonary aspergillosis (CPA). Three single nucleotide polymorphisms (SNPs; rs2305619, rs3816527, and rs1840680) in the PTX3 gene were detected by polymerase chain reaction direct sequencing, and their associations with aspergillosis were evaluated. Plasma PTX3 levels were measured by enzyme-linked immunosorbent assay. Results Three SNPs were consistent with Hardy-Weinberg equilibrium. The recessive model for SNP rs1840680 suggests that the AA homozygote is associated with a higher risk for pulmonary aspergillosis (both IPA and CPA) in COPD patients. However, for genotypic and allele frequencies of rs2305619 and rs3816527, no association was observed. The distribution of haplotypes (rs2305619 and rs3816527) also showed no significant differences. Plasma PTX3 levels significantly increased in IPA patients. In the IPA group, the rs1840680 AA genotype subjects showed lower plasma PTX3 levels than those harboring AG and GG genotypes. Conclusions This study showed a significant association between PTX3 rs1840680 polymorphisms and the susceptibility of pulmonary aspergillosis in patients with COPD. Plasma PTX3 levels may be potentially used as indicators for the diagnosis of IPA in COPD patients.
Collapse
Affiliation(s)
- Qian He
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southern Medical University, Guangzhou
| | - Hongxing Li
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southern Medical University, Guangzhou
| | - Yuwen Rui
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lulu Liu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Binchan He
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yi Shi
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southern Medical University, Guangzhou
| | - Xin Su
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southern Medical University, Guangzhou.,Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
16
|
Rathore M, Girard C, Ohanna M, Tichet M, Ben Jouira R, Garcia E, Larbret F, Gesson M, Audebert S, Lacour JP, Montaudié H, Prod'Homme V, Tartare-Deckert S, Deckert M. Cancer cell-derived long pentraxin 3 (PTX3) promotes melanoma migration through a toll-like receptor 4 (TLR4)/NF-κB signaling pathway. Oncogene 2019; 38:5873-5889. [PMID: 31253871 DOI: 10.1038/s41388-019-0848-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 03/30/2019] [Accepted: 04/28/2019] [Indexed: 01/23/2023]
Abstract
Cutaneous melanoma is one of the most aggressive cancers characterized by a high plasticity, a propensity for metastasis, and drug resistance. Melanomas are composed of phenotypically diverse subpopulations of tumor cells with heterogeneous molecular profiles that reflect intrinsic invasive abilities. In an attempt to identify novel factors of the melanoma invasive cell state, we previously investigated the nature of the invasive secretome by using a comparative proteomic approach. Here, we have extended this analysis to show that PTX3, an acute phase inflammatory glycoprotein, is one such factor secreted by invasive melanoma to promote tumor cell invasiveness. Elevated PTX3 production was observed in the population of MITFlow invasive cells but not in the population of MITFhigh differentiated melanoma cells. Consistently, MITF knockdown increased PTX3 expression in MITFhigh proliferative and poorly invasive cells. High levels of PTX3 were found in tissues and blood of metastatic melanoma patients, and in BRAF inhibitor-resistant melanoma cells displaying a mesenchymal invasive MITFlow phenotype. Genetic silencing of PTX3 in invasive melanoma cells dramatically impaired migration and invasion in vitro and in experimental lung extravasation assay in xenografted mice. In contrast, addition of melanoma-derived or recombinant PTX3, or expression of PTX3 enhanced motility of low migratory cells. Mechanistically, autocrine production of PTX3 by melanoma cells triggered an IKK/NFκB signaling pathway that promotes migration, invasion, and expression of the EMT factor TWIST1. Finally, we found that TLR4 and MYD88 knockdown inhibited PTX3-induced melanoma cell migration, suggesting that PTX3 functions through a TLR4-dependent pathway. Our work reveals that tumor-derived PTX3 contributes to melanoma cell invasion via targetable inflammation-related pathways. In addition to providing new insights into the biology of melanoma invasive behavior, this study underscores the notion that secreted PTX3 represents a potential biomarker and therapeutic target in a subpopulation of MITFlow invasive and/or refractory melanoma.
Collapse
Affiliation(s)
- M Rathore
- Université Côte d'Azur, INSERM, C3M, Nice, France
- Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - C Girard
- Université Côte d'Azur, INSERM, C3M, Nice, France
- Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - M Ohanna
- Université Côte d'Azur, INSERM, C3M, Nice, France
- Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - M Tichet
- Université Côte d'Azur, INSERM, C3M, Nice, France
- Laboratory of Translational Oncology, ISREC, EPFL, Lausanne, Switzerland
| | - R Ben Jouira
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | - E Garcia
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | - F Larbret
- Université Côte d'Azur, INSERM, C3M, Nice, France
- Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - M Gesson
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | - S Audebert
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - J-P Lacour
- Université Côte d'Azur, CHU Nice, Nice, France
| | - H Montaudié
- Université Côte d'Azur, CHU Nice, Nice, France
| | - V Prod'Homme
- Université Côte d'Azur, INSERM, C3M, Nice, France
- Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - S Tartare-Deckert
- Université Côte d'Azur, INSERM, C3M, Nice, France.
- Equipe labellisée Ligue Contre le Cancer 2016, Nice, France.
| | - M Deckert
- Université Côte d'Azur, INSERM, C3M, Nice, France.
- Equipe labellisée Ligue Contre le Cancer 2016, Nice, France.
| |
Collapse
|
17
|
Antunes D, Cunha C, Carvalho A. Genetic Regulation of the Host-Fungus Interaction in the Pathogenesis of Aspergillosis. CURRENT FUNGAL INFECTION REPORTS 2019. [DOI: 10.1007/s12281-019-00344-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
18
|
Mirkov I, Popov Aleksandrov A, Lazovic B, Glamoclija J, Kataranovski M. Usefulness of animal models of aspergillosis in studying immunity against Aspergillus infections. J Mycol Med 2019; 29:84-96. [DOI: 10.1016/j.mycmed.2019.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/28/2018] [Accepted: 01/14/2019] [Indexed: 01/08/2023]
|
19
|
Cunha C, Carvalho A. Genetic defects in fungal recognition and susceptibility to invasive pulmonary aspergillosis. Med Mycol 2019; 57:S211-S218. [PMID: 30816966 DOI: 10.1093/mmy/myy057] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/04/2018] [Accepted: 07/06/2018] [Indexed: 12/13/2022] Open
Abstract
The interindividual variability in the onset and clinical course of invasive pulmonary aspergillosis (IPA) raises fundamental questions about its actual pathogenesis. Clinical and epidemiological studies have reported only a few examples of monogenic defects, however an expanding number of common polymorphisms associated with IPA has been identified. Understanding how genetic variation regulates the immune response to Aspergillus provides critical insights into the human immunobiology of IPA by pinpointing directly relevant immune molecules and interacting pathways. Most of the genetic defects reported to increase susceptibility to infection were described or suggested to impair fungal recognition by the innate immune system. In this review, we discuss the contribution of host genetic variation in pattern recognition receptors to the development of IPA. An improved understanding of the molecular and cellular processes that regulate human susceptibility to IPA is ultimately expected to pave the way toward personalized medical interventions based on host-directed risk stratification and individualized immunotherapy.
Collapse
Affiliation(s)
- Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
20
|
Ugajin M, Kani H. A case of invasive pulmonary aspergillosis during treatment for acute exacerbation of interstitial lung disease. Infect Dis Rep 2018; 10:7785. [PMID: 30662692 PMCID: PMC6315312 DOI: 10.4081/idr.2018.7785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/09/2018] [Indexed: 11/23/2022] Open
Abstract
Prolonged immunosuppressive therapy is a risk factor for invasive pulmonary aspergillosis. We report a case of a 79-yearold man who underwent immunosuppressive therapy with methylprednisolone and cyclosporine for an acute exacerbation of interstitial lung disease. Ten days after initiation of immunosuppressive therapy, the patient reported night sweats and purulent sputum, and chest computed tomography scan revealed consolidation. He was diagnosed with invasive pulmonary aspergillosis, and required vasopressor support with oxygen therapy. After the administration of voriconazole and the modulation of immunosuppressive therapy, his condition improved. Short-term immunosuppressive therapy can also induce invasive pulmonary aspergillosis.
Collapse
Affiliation(s)
| | - Hisanori Kani
- Thoracic Surgery, Nagoya Tokushukai General Hospital, Japan
| |
Collapse
|
21
|
Garlanda C, Bottazzi B, Magrini E, Inforzato A, Mantovani A. PTX3, a Humoral Pattern Recognition Molecule, in Innate Immunity, Tissue Repair, and Cancer. Physiol Rev 2018; 98:623-639. [PMID: 29412047 DOI: 10.1152/physrev.00016.2017] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Innate immunity includes a cellular and a humoral arm. PTX3 is a fluid-phase pattern recognition molecule conserved in evolution which acts as a key component of humoral innate immunity in infections of fungal, bacterial, and viral origin. PTX3 binds conserved microbial structures and self-components under conditions of inflammation and activates effector functions (complement, phagocytosis). Moreover, it has a complex regulatory role in inflammation, such as ischemia/reperfusion injury and cancer-related inflammation, as well as in extracellular matrix organization and remodeling, with profound implications in physiology and pathology. Finally, PTX3 acts as an extrinsic oncosuppressor gene by taming tumor-promoting inflammation in murine and selected human tumors. Thus evidence suggests that PTX3 is a key homeostatic component at the crossroad of innate immunity, inflammation, tissue repair, and cancer. Dissecting the complexity of PTX3 pathophysiology and human genetics paves the way to diagnostic and therapeutic exploitation.
Collapse
Affiliation(s)
- Cecilia Garlanda
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Barbara Bottazzi
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Elena Magrini
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Antonio Inforzato
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
22
|
Sam QH, Yew WS, Seneviratne CJ, Chang MW, Chai LYA. Immunomodulation as Therapy for Fungal Infection: Are We Closer? Front Microbiol 2018; 9:1612. [PMID: 30090091 PMCID: PMC6068232 DOI: 10.3389/fmicb.2018.01612] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/28/2018] [Indexed: 12/20/2022] Open
Abstract
Invasive fungal disease (IFD) causes significant morbidity in immunocompromised patients due to their weakened immune system. Immunomodulatory therapy, in synergy with existing antifungal therapy, is an attractive option to enhance their immune system and aid clearance of these opportunistic pathogens. From a scientific and clinical perspective, we explore the immunotherapeutic options to augment standard antifungal drugs for patients with an IFD. We discuss the range of immunomodulatory therapies being considered in IFD - from cytokines, including G-CSF, GM-CSF, M-CSF, IFN-γ, and cytokine agonists, to cellular therapies, consisting of granulocyte transfusion, adoptive T-cell, CAR T-cell, natural killer cell therapies, and monoclonal antibodies. Adjunct pharmaceutical agents which augment the immunity are also being considered. Lastly, we explore the likelihood of the use of probiotics and manipulation of the microbiome/mycobiome to enhance IFD treatment outcomes.
Collapse
Affiliation(s)
- Qi Hui Sam
- Division of Infectious Diseases, University Medicine Cluster – National University Health System, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Wen Shan Yew
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | | | - Matthew Wook Chang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Louis Yi Ann Chai
- Division of Infectious Diseases, University Medicine Cluster – National University Health System, Singapore, Singapore
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National University Cancer Institute, Singapore, Singapore
| |
Collapse
|
23
|
The role of neutrophils in host defense against invasive fungal infections. CURRENT CLINICAL MICROBIOLOGY REPORTS 2018; 5:181-189. [PMID: 31552161 DOI: 10.1007/s40588-018-0098-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Purpose of Review Invasive fungal infections caused by the commensal yeast Candida and the ubiquitous, inhaled mold Aspergillus have emerged as major causes of morbidity and mortality in critically ill and immunosuppressed patient populations. Here, we review how neutrophils contribute to effective immunity against these infections. Recent Findings Studies in mouse models of invasive candidiasis and aspergillosis, and observations in hematological patients with chemotherapy-induced neutropenia and in patients with primary immunodeficiency disorders that manifest with these infections have highlighted the critical role of neutrophils and have identified key immune factors that promote neutrophil-mediated effective host defense against invasive fungal disease. Summary Neutrophils are crucial in host protection against invasive candidiasis and aspergillosis. Recent advances in our understanding of the molecular cues that mediate protective neutrophil recruitment and effector function against these infections hold promise for developing immune-based strategies to improve the outcomes of affected patients.
Collapse
|
24
|
Aerobic fitness alters the capacity of mononuclear cells to produce pentraxin 3 following maximal exercise. Eur J Appl Physiol 2018; 118:1515-1526. [PMID: 29748720 DOI: 10.1007/s00421-018-3882-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/01/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Pentraxin 3 (PTX3) is a vital regulator of innate immune function. Although plasma PTX3 concentrations are elevated with aerobic fitness, the cellular functions of PTX3 remain unknown in aerobically trained and untrained subjects. METHODS Thirty individuals (aerobically trained = 15 and untrained = 15) participated in a maximal exercise protocol to examine ex vivo PTX3 production from isolated peripheral blood mononuclear cells (PBMCs) exposed to LPS or palmitate. The capacity of PTX3 to stimulate inflammatory cytokine production ex vivo was also examined. RESULTS Elevated plasma PTX3 concentrations prior to exercise were positively associated with the percent change (pre to post exercise) in plasma PTX3 concentrations in all subjects, independent of cardiorespiratory fitness (VO2max). In addition, elevated plasma PTX3 concentrations in aerobically trained subjects at rest predicted changes in the LPS- and palmitate-stimulated PTX3 production from isolated PBMCs following acute exercise. In response to PTX3 simulation, the capacity of PBMCs to produce the anti-inflammatory cytokine IL-10 was decreased following acute exercise in all subject (no changes in IL-6, TGF-β1, and TNF-α observed). However, the percent change in IL-6 production was positively associated with VO2max in all subjects, and in aerobically trained subjects only, positively associated with elevated plasma PTX3 concentrations at rest and in response to acute exercise. CONCLUSION These results suggest that aerobic training enhances the utilization of plasma PTX3 concentrations to predict the capacity of mononuclear cells to produce PTX3, and potentially, its reciprocal role of PTX3 as an initiator of the innate immune response following maximal exercise.
Collapse
|
25
|
Grčević D, Sironi M, Valentino S, Deban L, Cvija H, Inforzato A, Kovačić N, Katavić V, Kelava T, Kalajzić I, Mantovani A, Bottazzi B. The Long Pentraxin 3 Plays a Role in Bone Turnover and Repair. Front Immunol 2018; 9:417. [PMID: 29556234 PMCID: PMC5845433 DOI: 10.3389/fimmu.2018.00417] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 02/15/2018] [Indexed: 01/04/2023] Open
Abstract
Pentraxin 3 (PTX3) is an inflammatory mediator acting as a fluid-phase pattern recognition molecule and playing an essential role in innate immunity and matrix remodeling. Inflammatory mediators also contribute to skeletal homeostasis, operating at multiple levels in physiological and pathological conditions. This study was designed to investigate the role of PTX3 in physiological skeletal remodeling and bone healing. Micro-computed tomography (μCT) and bone histomorphometry of distal femur showed that PTX3 gene-targeted female and male mice (ptx3−/−) had lower trabecular bone volume than their wild-type (ptx3+/+) littermates (BV/TV by μCT: 3.50 ± 1.31 vs 6.09 ± 1.17 for females, p < 0.0001; BV/TV 9.06 ± 1.89 vs 10.47 ± 1.97 for males, p = 0.0435). In addition, μCT revealed lower trabecular bone volume in second lumbar vertebra of ptx3−/− mice. PTX3 was increasingly expressed during osteoblast maturation in vitro and was able to reverse the negative effect of fibroblast growth factor 2 (FGF2) on osteoblast differentiation. This effect was specific for the N-terminal domain of PTX3 that contains the FGF2-binding site. By using the closed transversal tibial fracture model, we found that ptx3−/− female mice formed significantly less mineralized callus during the anabolic phase following fracture injury compared to ptx3+/+ mice (BV/TV 17.05 ± 4.59 vs 20.47 ± 3.32, p = 0.0195). Non-hematopoietic periosteal cells highly upregulated PTX3 expression during the initial phase of fracture healing, particularly CD51+ and αSma+ osteoprogenitor subsets, and callus tissue exhibited concomitant expression of PTX3 and FGF2 around the fracture site. Thus, PTX3 supports maintenance of the bone mass possibly by inhibiting FGF2 and its negative impact on bone formation. Moreover, PTX3 enables timely occurring sequence of callus mineralization after bone fracture injury. These results indicate that PTX3 plays an important role in bone homeostasis and in proper matrix mineralization during fracture repair, a reflection of the function of this molecule in tissue homeostasis and repair.
Collapse
Affiliation(s)
- Danka Grčević
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Marina Sironi
- Humanitas Clinical and Research Center, Milan, Italy
| | | | - Livija Deban
- Humanitas Clinical and Research Center, Milan, Italy.,Oxford BioTherapeutics Ltd., Abingdon, United Kingdom
| | - Hrvoje Cvija
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Antonio Inforzato
- Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Nataša Kovačić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vedran Katavić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Tomislav Kelava
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivo Kalajzić
- Department of Reconstructive Sciences, School of Dental Medicine, UConn Health, Farmingam, CT, United States
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
26
|
Dander E, De Lorenzo P, Bottazzi B, Quarello P, Vinci P, Balduzzi A, Masciocchi F, Bonanomi S, Cappuzzello C, Prunotto G, Pavan F, Pasqualini F, Sironi M, Cuccovillo I, Leone R, Salvatori G, Parma M, Terruzzi E, Pagni F, Locatelli F, Mantovani A, Fagioli F, Biondi A, Garlanda C, Valsecchi MG, Rovelli A, D'Amico G. Pentraxin 3 plasma levels at graft-versus-host disease onset predict disease severity and response to therapy in children given haematopoietic stem cell transplantation. Oncotarget 2018; 7:82123-82138. [PMID: 27893415 PMCID: PMC5347680 DOI: 10.18632/oncotarget.13488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/15/2016] [Indexed: 12/05/2022] Open
Abstract
Acute Graft-versus-Host Disease (GvHD) remains a major complication of allogeneic haematopoietic stem cell transplantation, with a significant proportion of patients failing to respond to first-line systemic corticosteroids. Reliable biomarkers predicting disease severity and response to treatment are warranted to improve its management. Thus, we sought to determine whether pentraxin 3 (PTX3), an acute-phase protein produced locally at the site of inflammation, could represent a novel acute GvHD biomarker. Using a murine model of the disease, we found increased PTX3 plasma levels after irradiation and at GvHD onset. Similarly, plasma PTX3 was enhanced in 115 pediatric patients on day of transplantation, likely due to conditioning, and at GvHD onset in patients experiencing clinical symptoms of the disease. PTX3 was also found increased in skin and colon biopsies from patients with active disease. Furthermore, PTX3 plasma levels at GvHD onset were predictive of disease outcome since they resulted significantly higher in both severe and therapy-unresponsive patients. Multiple injections of rhPTX3 in the murine model of GvHD did not influence the disease course. Taken together, our results indicate that PTX3 constitutes a biomarker of GvHD severity and therapy response useful to tailor treatment intensity according to early risk-stratification of GvHD patients.
Collapse
Affiliation(s)
- Erica Dander
- "M. Tettamanti" Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Paola De Lorenzo
- Center of Biostatistics for Clinical Epidemiology, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Barbara Bottazzi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - Humanitas Clinical and Research Center, Rozzano, Italy
| | - Paola Quarello
- Pediatric Onco-Haematology, City of Science and Health of Turin, Regina Margherita Children's Hospital, Torino, Italy
| | - Paola Vinci
- "M. Tettamanti" Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Adriana Balduzzi
- Clinica Pediatrica Ospedale S. Gerardo, Fondazione MBBM, University of Milano-Bicocca, Monza, Italy
| | - Francesca Masciocchi
- Clinica Pediatrica Ospedale S. Gerardo, Fondazione MBBM, University of Milano-Bicocca, Monza, Italy
| | - Sonia Bonanomi
- Clinica Pediatrica Ospedale S. Gerardo, Fondazione MBBM, University of Milano-Bicocca, Monza, Italy
| | - Claudia Cappuzzello
- "M. Tettamanti" Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Giulia Prunotto
- Clinica Pediatrica Ospedale S. Gerardo, Fondazione MBBM, University of Milano-Bicocca, Monza, Italy
| | - Fabio Pavan
- Clinica Pediatrica Ospedale S. Gerardo, Fondazione MBBM, University of Milano-Bicocca, Monza, Italy
| | - Fabio Pasqualini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - Humanitas Clinical and Research Center, Rozzano, Italy
| | - Marina Sironi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - Humanitas Clinical and Research Center, Rozzano, Italy
| | - Ivan Cuccovillo
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - Humanitas Clinical and Research Center, Rozzano, Italy
| | - Roberto Leone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - Humanitas Clinical and Research Center, Rozzano, Italy
| | - Giovanni Salvatori
- Sigma-tau S.p.A., Department of R&D, Biotechnology, San Gerardo Hospital, Monza, Italy
| | - Matteo Parma
- Haematology Division and BMT Unit, San Gerardo Hospital, Monza, Italy
| | | | - Fabio Pagni
- Department of Surgery and Interdisciplinary Medicine, University Milano-Bicocca, Section of Pathology, San Gerardo Hospital, Monza, Italy
| | - Franco Locatelli
- Department of Pediatric Haematology-Oncology, IRCCS, Bambino Gesù Children Hospital, Roma-Department of Pediatric Science, University of Pavia, Pavia, Italy
| | - Alberto Mantovani
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - Humanitas Clinical and Research Center, Rozzano, Italy.,Humanitas University, Rozzano, Italy
| | - Franca Fagioli
- Pediatric Onco-Haematology, City of Science and Health of Turin, Regina Margherita Children's Hospital, Torino, Italy
| | - Andrea Biondi
- "M. Tettamanti" Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy.,Clinica Pediatrica Ospedale S. Gerardo, Fondazione MBBM, University of Milano-Bicocca, Monza, Italy
| | - Cecilia Garlanda
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - Humanitas Clinical and Research Center, Rozzano, Italy
| | - Maria Grazia Valsecchi
- Center of Biostatistics for Clinical Epidemiology, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Attilio Rovelli
- Clinica Pediatrica Ospedale S. Gerardo, Fondazione MBBM, University of Milano-Bicocca, Monza, Italy
| | - Giovanna D'Amico
- "M. Tettamanti" Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
27
|
Immune responses to invasive aspergillosis: new understanding and therapeutic opportunities. Curr Opin Infect Dis 2018; 30:364-371. [PMID: 28509673 DOI: 10.1097/qco.0000000000000381] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Invasive aspergillosis is a worldwide disease that primarily affects immune-compromised patients, agricultural workers with corneal abrasions, individuals with structural lung disease, and patients with primary immune deficiency. The critical function of the immune system is to prevent the germination of airborne conidia into tissue-invasive hyphae. This review covers recent advances that shape our understanding of anti-Aspergillus immunity at the molecular and cellular level. RECENT FINDINGS Host defense against conidia and hyphae occurs via distinct molecular mechanisms that involve intracellular and extracellular killing pathways, as well as cooperation between different myeloid cell subsets. The strength and efficacy of the host response is shaped by the tissue microenvironment. In preclinical models of disease, host immune augmentation strategies have yielded benefits, yet translating these insights into therapeutic strategies in humans remains challenging. SUMMARY Although advances in early diagnostic strategies and in antifungal drugs have ameliorated clinical outcomes of invasive aspergillosis, further improvements depend on gaining deeper insight into and translating advances in anti-Aspergillus immunity.
Collapse
|
28
|
Campos CF, van de Veerdonk FL, Gonçalves SM, Cunha C, Netea MG, Carvalho A. Host Genetic Signatures of Susceptibility to Fungal Disease. Curr Top Microbiol Immunol 2018; 422:237-263. [PMID: 30043341 DOI: 10.1007/82_2018_113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Our relative inability to predict the development of fungal disease and its clinical outcome raises fundamental questions about its actual pathogenesis. Several clinical risk factors are described to predispose to fungal disease, particularly in immunocompromised and severely ill patients. However, these alone do not entirely explain why, under comparable clinical conditions, only some patients develop infection. Recent clinical and epidemiological studies have reported an expanding number of monogenic defects and common polymorphisms associated with fungal disease. By directly implicating genetic variation in the functional regulation of immune mediators and interacting pathways, these studies have provided critical insights into the human immunobiology of fungal disease. Most of the common genetic defects reported were described or suggested to impair fungal recognition by the innate immune system. Here, we review common genetic variation in pattern recognition receptors and its impact on the immune response against the two major fungal pathogens Candida albicans and Aspergillus fumigatus. In addition, we discuss potential strategies and opportunities for the clinical translation of genetic information in the field of medical mycology. These approaches are expected to transfigure current clinical practice by unleashing an unprecedented ability to personalize prophylaxis, therapy and monitoring for fungal disease.
Collapse
Affiliation(s)
- Cláudia F Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Frank L van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboudumc, Nijmegen, The Netherlands
| | - Samuel M Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboudumc, Nijmegen, The Netherlands
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
29
|
Perea L, Coll M, Sanjurjo L, Blaya D, Taghdouini AE, Rodrigo-Torres D, Altamirano J, Graupera I, Aguilar-Bravo B, Llopis M, Vallverdú J, Caballeria J, van Grunsven LA, Sarrias MR, Ginès P, Sancho-Bru P. Pentraxin-3 modulates lipopolysaccharide-induced inflammatory response and attenuates liver injury. Hepatology 2017; 66:953-968. [PMID: 28422322 PMCID: PMC5570620 DOI: 10.1002/hep.29215] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 04/04/2017] [Accepted: 04/13/2017] [Indexed: 12/13/2022]
Abstract
Acute-on-chronic liver injury is characterized by an important inflammatory response frequently associated with endotoxemia. In this context, acute-phase proteins such as Pentraxin-3 (PTX3) are released; however, little is known about their role in chronic liver disease. The aim of this study was to elucidate the role of PTX3 in liver injury. The role of PTX3 was evaluated in cultured human cells, liver tissue slices, and mice with acute-on-chronic liver injury. PTX3 expression was assessed in tissue and serum samples from 54 patients with alcoholic hepatitis. PTX3 expression was up-regulated in animal models of liver injury and strongly induced by lipopolysaccharide (LPS). Liver cell fractionation showed that macrophages and activated hepatic stellate cells were the main cell types expressing PTX3 in liver injury. Ex vivo and in vivo studies showed that PTX3 treatment attenuated LPS-induced liver injury, inflammation, and cell recruitment. Mechanistically, PTX3 mediated the hepatic stellate cell wound-healing response. Moreover, PTX3 modulated LPS-induced inflammation in human primary liver macrophages and peripheral monocytes by enhancing a TIR domain-containing adapter-inducing interferon-dependent response and favoring a macrophage interleukin-10-like phenotype. Additionally, hepatic and plasma PTX3 levels were increased in patients with alcoholic hepatitis, a prototypic acute-on-chronic condition; and its expression correlated with disease severity scores, endotoxemia, infections, and short-term mortality, thus suggesting that expression of PTX3 found in patients could be a counterregulatory response to injury. CONCLUSION Experimental and human evidence suggests that, in addition to being a potential biomarker for alcoholic hepatitis, PTX3 participates in the wound-healing response and attenuates LPS-induced liver injury and inflammation; therefore, administration of PTX3 could be a promising therapeutic strategy in acute-on-chronic conditions, particularly those associated with endotoxemia. (Hepatology 2017;66:953-968).
Collapse
Affiliation(s)
- Luis Perea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mar Coll
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Lucia Sanjurjo
- Innate Immunity Group, Health Sciences Research Institute Germans Trias i Pujol, Badalona, Spain
- Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Delia Blaya
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Adil El Taghdouini
- Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Daniel Rodrigo-Torres
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - José Altamirano
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Isabel Graupera
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Beatriz Aguilar-Bravo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marta Llopis
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Julia Vallverdú
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Joan Caballeria
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Liver Unit, Hospital Clínic, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Leo A. van Grunsven
- Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Maria-Rosa Sarrias
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Innate Immunity Group, Health Sciences Research Institute Germans Trias i Pujol, Badalona, Spain
| | - Pere Ginès
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Liver Unit, Hospital Clínic, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Pau Sancho-Bru
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| |
Collapse
|
30
|
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
|
31
|
Abstract
Pathogenic fungi cause a wide range of syndromes in immune-competent and immune-compromised individuals, with life-threatening disease primarily seen in humans with HIV/AIDS and in patients receiving immunosuppressive therapies for cancer, autoimmunity, and end-organ failure. The discovery that specific primary immune deficiencies manifest with fungal infections and the development of animal models of mucosal and invasive mycoses have facilitated insight into fungus-specific recognition, signaling, effector pathways, and adaptive immune responses. Progress in deciphering the molecular and cellular basis of immunity against fungi is guiding preclinical studies into vaccine and immune reconstitution strategies for vulnerable patient groups. Furthermore, recent work has begun to address the role of endogenous fungal communities in human health and disease. In this review, we summarize a contemporary understanding of protective immunity against fungi.
Collapse
Affiliation(s)
- Michail S Lionakis
- Fungal Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Iliyan D Iliev
- Jill Roberts Institute for Research in IBD, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Tobias M Hohl
- Infectious Disease Service, Department of Medicine, and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
32
|
Desoubeaux G, Cray C. Rodent Models of Invasive Aspergillosis due to Aspergillus fumigatus: Still a Long Path toward Standardization. Front Microbiol 2017; 8:841. [PMID: 28559881 PMCID: PMC5432554 DOI: 10.3389/fmicb.2017.00841] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 04/24/2017] [Indexed: 01/09/2023] Open
Abstract
Invasive aspergillosis has been studied in laboratory by the means of plethora of distinct animal models. They were developed to address pathophysiology, therapy, diagnosis, or miscellaneous other concerns associated. However, there are great discrepancies regarding all the experimental variables of animal models, and a thorough focus on them is needed. This systematic review completed a comprehensive bibliographic analysis specifically-based on the technical features of rodent models infected with Aspergillus fumigatus. Out the 800 articles reviewed, it was shown that mice remained the preferred model (85.8% of the referenced reports), above rats (10.8%), and guinea pigs (3.8%). Three quarters of the models involved immunocompromised status, mainly by steroids (44.4%) and/or alkylating drugs (42.9%), but only 27.7% were reported to receive antibiotic prophylaxis to prevent from bacterial infection. Injection of spores (30.0%) and inhalation/deposition into respiratory airways (66.9%) were the most used routes for experimental inoculation. Overall, more than 230 distinct A. fumigatus strains were used in models. Of all the published studies, 18.4% did not mention usage of any diagnostic tool, like histopathology or mycological culture, to control correct implementation of the disease and to measure outcome. In light of these findings, a consensus discussion should be engaged to establish a minimum standardization, although this may not be consistently suitable for addressing all the specific aspects of invasive aspergillosis.
Collapse
Affiliation(s)
- Guillaume Desoubeaux
- Division of Comparative Pathology, Department of Pathology and Laboratory Medicine, Miller School of Medicine, University of MiamiMiami, FL, USA.,Service de Parasitologie-Mycologie-Médecine tropicale, Centre Hospitalier Universitaire de ToursTours, France.,Centre d'Etude des Pathologies Respiratoires (CEPR) Institut National de la Santé et de la Recherche Médicale U1100/Équipe 3, Université François-RabelaisTours, France
| | - Carolyn Cray
- Division of Comparative Pathology, Department of Pathology and Laboratory Medicine, Miller School of Medicine, University of MiamiMiami, FL, USA
| |
Collapse
|
33
|
Slusher AL, Shibata Y, Whitehurst M, Maharaj A, Quiles JM, Huang CJ. Exercise reduced pentraxin 3 levels produced by endotoxin-stimulated human peripheral blood mononuclear cells in obese individuals. Exp Biol Med (Maywood) 2017; 242:1279-1286. [PMID: 28440716 DOI: 10.1177/1535370217706963] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The purpose of this study was to determine whether obesity would reduce the capacity of peripheral blood mononuclear cells (PBMCs) to produce the anti-inflammatory protein pentraxin 3 (PTX3) in response to ex vivo stimulation with lipopolysaccharide (LPS), and if acute aerobic exercise would enhance this PTX3 production capacity. In addition, the inter-relationships of LPS-induced PTX3 with the inflammatory cytokines (interleukin 6 [IL-6], IL-10, and tumor necrosis factor alpha) were examined. Twenty-one healthy subjects (10 obese and 11 normal-weight) performed an acute bout of aerobic exercise at 75% VO2max. The capacity of PBMCs to produce PTX3 ex vivo following LPS stimulation was the same in obese and normal-weight subjects at rest, and decreased equally in both subject groups following acute aerobic exercise. This is in contrast to plasma PTX3, which is lower in obese subjects at rest and increased equally in both obese and normal-weight subjects following exercise. In addition, ex vivo PTX3 production was positively associated with IL-6 and IL-10 in response to acute aerobic exercise ( r = 0.686, P = 0.020; r = 0.744, P = 0.009, respectively) in normal-weight, but not in obese individuals ( r = 0.429, P = 0.249; r = 0.453, P = 0.189, respectively). These findings indicate that concentrations of PTX3 observed in plasma are relatively independent of those produced by PBMCs ex vivo and the mechanisms associated with PTX3-mediated anti-inflammatory signaling may differ during obesity. Impact statement Our laboratory has previously demonstrated that obese individuals present with lower plasma concentrations of the anti-inflammatory protein pentraxin 3 (PTX3), whereas acute aerobic exercise increases plasma PTX3 levels similarly compared to normal-weight individuals. As a follow-up, the present study demonstrates that PBMCs isolated from obese and normal-weight individuals produce comparable amounts of PTX3 ex vivo in response to lipopolysaccharide (LPS). Furthermore, given that acute aerobic exercise reduced the ex vivo production of PTX3 in both groups, our results clearly indicate that plasma PTX3 levels are relatively independent of those produced by PBMCs ex vivo. In addition, our findings suggest that the mechanisms associated with PTX3-mediated production of the anti-inflammatory cytokine interleukin 10 may be impaired in obese individuals, and thus provides a key finding necessary for the elucidation of PTX3's role in the mediation of anti-inflammatory profiles and the subsequent amelioration of inflammatory disease during obesity.
Collapse
Affiliation(s)
- Aaron L Slusher
- 1 Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA.,2 Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Yoshimi Shibata
- 3 Department of Biomedical Science, College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Michael Whitehurst
- 1 Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Arun Maharaj
- 1 Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Justin M Quiles
- 1 Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Chun-Jung Huang
- 1 Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
34
|
The Potential Role of Aerobic Exercise-Induced Pentraxin 3 on Obesity-Related Inflammation and Metabolic Dysregulation. Mediators Inflamm 2017; 2017:1092738. [PMID: 28400677 PMCID: PMC5376480 DOI: 10.1155/2017/1092738] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 02/08/2017] [Accepted: 03/06/2017] [Indexed: 01/01/2023] Open
Abstract
Obesity is defined as the excess accumulation of intra-abdominal body fat, resulting in a state of chronic, low-grade proinflammation that can directly contribute to the development of insulin resistance. Pentraxin 3 (PTX3) is an acute-phase protein that is expressed by a variety of tissue and cell sources and provides an anti-inflammatory property to downregulate the production of proinflammatory cytokines, in particular interleukin-1 beta and tumor necrosis factor alpha. Although PTX3 may therapeutically aid in altering the proinflammatory milieu in obese individuals, and despite elevated expression of PTX3 mRNA observed in adipose tissue, the circulating level of PTX3 is reduced with obesity. Interestingly, aerobic activity has been demonstrated to elevate PTX3 levels. Therefore, the purpose of this review is to discuss the therapeutic potential of PTX3 to positively regulate obesity-related inflammation and discuss the proposition for utilizing aerobic exercise as a nonpharmacological anti-inflammatory treatment strategy to enhance circulating PTX3 concentrations in obese individuals.
Collapse
|
35
|
Zelante T, Wong AYW, Mencarelli A, Foo S, Zolezzi F, Lee B, Poidinger M, Ricciardi-Castagnoli P, Fric J. Impaired calcineurin signaling in myeloid cells results in downregulation of pentraxin-3 and increased susceptibility to aspergillosis. Mucosal Immunol 2017; 10:470-480. [PMID: 27301880 DOI: 10.1038/mi.2016.52] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 05/08/2016] [Indexed: 02/04/2023]
Abstract
Treatment of post-transplant patients with immunosuppressive drugs targeting the calcineurin-nuclear factor of activated T cells (NFAT) pathway, including cyclosporine A or tacrolimus, is commonly associated with a higher incidence of opportunistic infections, such as Aspergillus fumigatus, which can lead to severe life-threatening conditions. A component of the A. fumigatus cell wall, β-glucan, is recognized by dendritic cells (DCs) via the Dectin-1 receptor, triggering downstream signaling that leads to calcineurin-NFAT binding, NFAT translocation, and transcription of NFAT-regulated genes. Here, we address the question of whether calcineurin signaling in CD11c-expressing cells, such as DCs, has a specific role in the innate control of A. fumigatus. Impairment of calcineurin in CD11c-expressing cells (CD11ccrecnb1loxP) significantly increased susceptibility to systemic A. fumigatus infection and to intranasal infection in irradiated mice undergoing bone marrow transplant. Global expression profiling of bone marrow-derived DCs identified calcineurin-regulated processes in the immune response to infection, including expression of pentraxin-3, an important antifungal defense protein. These results suggest that calcineurin inhibition directly impairs important immunoprotective functions of myeloid cells, as shown by the higher susceptibility of CD11ccrecnbloxP mice in models of systemic and invasive pulmonary aspergillosis, including after allogeneic bone marrow transplantation. These findings are relevant to the clinical management of transplant patients with severe Aspergillus infections.
Collapse
Affiliation(s)
- T Zelante
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore.,Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - A Y W Wong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore.,National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - A Mencarelli
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore.,Emerging Infectious Diseases Programme, Duke-NUS, Singapore
| | - S Foo
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - F Zolezzi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - B Lee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - M Poidinger
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore
| | - P Ricciardi-Castagnoli
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - J Fric
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore.,Center for Translational Medicine, International Clinical Research Center, St Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
36
|
Garth JM, Steele C. Innate Lung Defense during Invasive Aspergillosis: New Mechanisms. J Innate Immun 2017; 9:271-280. [PMID: 28231567 DOI: 10.1159/000455125] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/15/2016] [Indexed: 12/19/2022] Open
Abstract
Invasive aspergillosis (IA) is one of the most difficult to treat and, consequently, one of the most lethal fungal infections known to man. Continued use of immunosuppressive agents during chemotherapy and organ transplantation often leads to the development of neutropenia, the primary risk factor for IA. However, IA is also becoming more appreciated in chronic diseases associated with corticosteroid therapy. The innate immune response to Aspergillus fumigatus, the primary agent in IA, plays a pivotal role in the recognition and elimination of organisms from the pulmonary system. This review highlights recent findings about innate host defense mechanisms, including novel aspects of innate cellular immunity and pathogen recognition, and the inflammatory mediators that control infection with A. fumigatus.
Collapse
Affiliation(s)
- Jaleesa M Garth
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | |
Collapse
|
37
|
Abstract
The innate immune system represents the first line of defense against pathogens and comprises both a cellular and a humoral arm. Fluid-phase pattern recognition molecules (PRMs), which include collectins, ficolins, and pentraxins, are key components of the humoral arm of innate immunity and are expressed by a variety of cells, including myeloid, epithelial, and endothelial cells, mainly in response to infectious and inflammatory conditions. Soluble PRMs share basic multifunctional properties including activation and regulation of the complement cascade, opsonization of pathogens and apoptotic cells, regulation of leukocyte extravasation, and fine-tuning of inflammation. Therefore, soluble PRMs are part of the immune response and retain antibody-like effector functions. Here, we will review the expression and general function of soluble PRMs, focusing our attention on the long pentraxin PTX3.
Collapse
|
38
|
Chorny A, Casas-Recasens S, Sintes J, Shan M, Polentarutti N, García-Escudero R, Walland AC, Yeiser JR, Cassis L, Carrillo J, Puga I, Cunha C, Bastos H, Rodrigues F, Lacerda JF, Morais A, Dieguez-Gonzalez R, Heeger PS, Salvatori G, Carvalho A, Garcia-Sastre A, Blander JM, Mantovani A, Garlanda C, Cerutti A. The soluble pattern recognition receptor PTX3 links humoral innate and adaptive immune responses by helping marginal zone B cells. J Exp Med 2016; 213:2167-85. [PMID: 27621420 PMCID: PMC5030794 DOI: 10.1084/jem.20150282] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 08/04/2016] [Indexed: 01/01/2023] Open
Abstract
Cerutti and collaborators show that the humoral arms of the innate and adaptive immune systems are functionally interconnected by pentraxin 3, a soluble pattern recognition receptor that couples innate immune recognition with antibody-inducing function. Pentraxin 3 (PTX3) is a fluid-phase pattern recognition receptor of the humoral innate immune system with ancestral antibody-like properties but unknown antibody-inducing function. In this study, we found binding of PTX3 to splenic marginal zone (MZ) B cells, an innate-like subset of antibody-producing lymphocytes strategically positioned at the interface between the circulation and the adaptive immune system. PTX3 was released by a subset of neutrophils that surrounded the splenic MZ and expressed an immune activation–related gene signature distinct from that of circulating neutrophils. Binding of PTX3 promoted homeostatic production of IgM and class-switched IgG antibodies to microbial capsular polysaccharides, which decreased in PTX3-deficient mice and humans. In addition, PTX3 increased IgM and IgG production after infection with blood-borne encapsulated bacteria or immunization with bacterial carbohydrates. This immunogenic effect stemmed from the activation of MZ B cells through a neutrophil-regulated pathway that elicited class switching and plasmablast expansion via a combination of T cell–independent and T cell–dependent signals. Thus, PTX3 may bridge the humoral arms of the innate and adaptive immune systems by serving as an endogenous adjuvant for MZ B cells. This property could be harnessed to develop more effective vaccines against encapsulated pathogens.
Collapse
Affiliation(s)
- Alejo Chorny
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Sandra Casas-Recasens
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jordi Sintes
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain
| | - Meimei Shan
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Nadia Polentarutti
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Clinical and Research Hospital, Rozzano, 20089 Milan, Italy
| | - Ramón García-Escudero
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales, y Tecnológicas, 28040 Madrid, Spain
| | - A Cooper Walland
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - John R Yeiser
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Linda Cassis
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain
| | - Jorge Carrillo
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain
| | - Irene Puga
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain
| | - Cristina Cunha
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal PT Government Associate Laboratory, Braga/Guimarães, Life and Health Sciences Research Institute /3B's, University of Minho, 4710-057 Braga, Portugal
| | - Hélder Bastos
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal PT Government Associate Laboratory, Braga/Guimarães, Life and Health Sciences Research Institute /3B's, University of Minho, 4710-057 Braga, Portugal Serviço de Pneumologia, Centro Hospitalar São João, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Fernando Rodrigues
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal PT Government Associate Laboratory, Braga/Guimarães, Life and Health Sciences Research Institute /3B's, University of Minho, 4710-057 Braga, Portugal
| | - João F Lacerda
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal Hospital de Santa Maria, 1649-035 Lisboa, Portugal
| | - António Morais
- Serviço de Pneumologia, Centro Hospitalar São João, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Rebeca Dieguez-Gonzalez
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Peter S Heeger
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | - Agostinho Carvalho
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal PT Government Associate Laboratory, Braga/Guimarães, Life and Health Sciences Research Institute /3B's, University of Minho, 4710-057 Braga, Portugal
| | - Adolfo Garcia-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - J Magarian Blander
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Alberto Mantovani
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Clinical and Research Hospital, Rozzano, 20089 Milan, Italy Humanitas University, Rozzano, 20089 Milan, Italy
| | - Cecilia Garlanda
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Clinical and Research Hospital, Rozzano, 20089 Milan, Italy Humanitas University, Rozzano, 20089 Milan, Italy
| | - Andrea Cerutti
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain Catalan Institute for Research and Advanced Studies, 08003 Barcelona, Spain
| |
Collapse
|
39
|
Lupiañez CB, Villaescusa MT, Carvalho A, Springer J, Lackner M, Sánchez-Maldonado JM, Canet LM, Cunha C, Segura-Catena J, Alcazar-Fuoli L, Solano C, Fianchi L, Pagano L, Potenza L, Aguado JM, Luppi M, Cuenca-Estrella M, Lass-Flörl C, Einsele H, Vázquez L, Ríos-Tamayo R, Loeffler J, Jurado M, Sainz J. Common Genetic Polymorphisms within NFκB-Related Genes and the Risk of Developing Invasive Aspergillosis. Front Microbiol 2016; 7:1243. [PMID: 27570521 PMCID: PMC4982195 DOI: 10.3389/fmicb.2016.01243] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/26/2016] [Indexed: 12/02/2022] Open
Abstract
Invasive Aspergillosis (IA) is an opportunistic infection caused by Aspergillus, a ubiquitously present airborne pathogenic mold. A growing number of studies suggest a major host genetic component in disease susceptibility. Here, we evaluated whether 14 single-nucleotide polymorphisms within NFκB1, NFκB2, RelA, RelB, Rel, and IRF4 genes influence the risk of IA in a population of 834 high-risk patients (157 IA and 677 non-IA) recruited through a collaborative effort involving the aspBIOmics consortium and four European clinical institutions. No significant overall associations between selected SNPs and the risk of IA were found in this large cohort. Although a hematopoietic stem cell transplantation (HSCT)-stratified analysis revealed that carriers of the IRF4rs12203592T/T genotype had a six-fold increased risk of developing the infection when compared with those carrying the C allele (ORREC = 6.24, 95%CI 1.25–31.2, P = 0.026), the association of this variant with IA risk did not reach significance at experiment-wide significant threshold. In addition, we found an association of the IRF4AATC and IRF4GGTC haplotypes (not including the IRF4rs12203592T risk allele) with a decreased risk of IA but the magnitude of the association was similar to the one observed in the single-SNP analysis, which indicated that the haplotypic effect on IA risk was likely due to the IRF4rs12203592 SNP. Finally, no evidence of significant interactions among the genetic markers tested and the risk of IA was found. These results suggest that the SNPs on the studied genes do not have a clinically relevant impact on the risk of developing IA.
Collapse
Affiliation(s)
- Carmen B Lupiañez
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS GranadaGranada, Spain; Hematology Department, Virgen de las Nieves University HospitalGranada, Spain
| | - María T Villaescusa
- Hematology Department, University Hospital of SalamancaSalamanca, Spain; Hematology Department, Jiménez Díaz FoundationMadrid, Spain
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of MinhoBraga, Portugal; ICVS/3B's - PT Government Associate LaboratoryBraga, Portugal
| | - Jan Springer
- Universitätsklinikum Würzburg, Medizinische Klinik II Würzburg, Germany
| | - Michaela Lackner
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck Innsbruck, Austria
| | - José M Sánchez-Maldonado
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada Granada, Spain
| | - Luz M Canet
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada Granada, Spain
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of MinhoBraga, Portugal; ICVS/3B's - PT Government Associate LaboratoryBraga, Portugal
| | - Juana Segura-Catena
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS GranadaGranada, Spain; Hematology Department, Virgen de las Nieves University HospitalGranada, Spain
| | - Laura Alcazar-Fuoli
- Mycology Reference Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III Madrid, Spain
| | - Carlos Solano
- Hematology Department, Clinic University Hospital of Valencia Valencia, Spain
| | - Luana Fianchi
- Istituto di Ematologia, Università Cattolica del S. Cuore Rome, Italy
| | - Livio Pagano
- Istituto di Ematologia, Università Cattolica del S. Cuore Rome, Italy
| | - Leonardo Potenza
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia AOU Policlinico, Modena, Italy
| | - José M Aguado
- Unit of Infectious Diseases, University Hospital 12 de Octubre, Research Institute of Hospital 12 de Octubre (i+12) Madrid, Spain
| | - Mario Luppi
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia AOU Policlinico, Modena, Italy
| | - Manuel Cuenca-Estrella
- Mycology Reference Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III Madrid, Spain
| | - Cornelia Lass-Flörl
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck Innsbruck, Austria
| | - Hermann Einsele
- Universitätsklinikum Würzburg, Medizinische Klinik II Würzburg, Germany
| | - Lourdes Vázquez
- Hematology Department, University Hospital of Salamanca Salamanca, Spain
| | | | - Rafael Ríos-Tamayo
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS GranadaGranada, Spain; Hematology Department, Virgen de las Nieves University HospitalGranada, Spain
| | - Jurgen Loeffler
- Universitätsklinikum Würzburg, Medizinische Klinik II Würzburg, Germany
| | - Manuel Jurado
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS GranadaGranada, Spain; Hematology Department, Virgen de las Nieves University HospitalGranada, Spain
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS GranadaGranada, Spain; Hematology Department, Virgen de las Nieves University HospitalGranada, Spain
| |
Collapse
|
40
|
Slusher AL, Mischo AB, Acevedo EO. Pentraxin 3 is an anti-inflammatory protein associated with lipid-induced interleukin 10 in vitro. Cytokine 2016; 86:36-40. [PMID: 27450429 DOI: 10.1016/j.cyto.2016.07.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 07/15/2016] [Accepted: 07/16/2016] [Indexed: 11/17/2022]
Abstract
UNLABELLED Pentraxin 3 (PTX3) is an acute phase protein expressed in response to pro-inflammatory stimuli during atherosclerosis. However, recent findings suggest that PTX3 is a counter-regulatory protein which enhances the anti-inflammatory response. OBJECTIVE Therefore, the capacity of PTX3 to alter the inflammatory milieu following in vitro stimulation of PBMCs with the pro-inflammatory lipid, palmitate, was examined. METHODS PBMCs from 17 healthy male donors were isolated and cultured under four separate conditions; 200μmol/L palmitate, a physiologically relevant concentration of PTX3, in combination (pal+PTX3), and an unstimulated time-course control. RESULTS Palmitate-induced production of the counter-regulatory protein PTX3 was positively associated with the production of the anti-inflammatory cytokine interleukin 10 (IL-10) following in vitro stimulation of human PBMCs. Furthermore, stimulation of PBMCs in vitro with 500pg/mL PTX3 elicited a significantly greater increase in IL-10 production compared to the palmitate stimulated conditions. However, PTX3 stimulation did not result in the production of the pro-inflammatory cytokines IL-1β, IL-6, and tumor necrosis factor alpha, and when combined with palmitate, did not alter the pro-inflammatory milieu from PBMCs in this study. CONCLUSION These findings provide evidence supporting the role of PTX3 as a mediator of the anti-inflammatory response in physiologically relevant conditions, and suggests that PTX3 counter regulates the development of atherosclerosis by enhancing the production of IL-10.
Collapse
Affiliation(s)
- Aaron L Slusher
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA, USA.
| | - Amanda B Mischo
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Edmund O Acevedo
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
41
|
Bidula S, Schelenz S. A Sweet Response to a Sour Situation: The Role of Soluble Pattern Recognition Receptors in the Innate Immune Response to Invasive Aspergillus fumigatus Infections. PLoS Pathog 2016; 12:e1005637. [PMID: 27415780 PMCID: PMC4945084 DOI: 10.1371/journal.ppat.1005637] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Stefan Bidula
- Aberdeen Fungal Group, School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Silke Schelenz
- Department of Microbiology, Royal Brompton Hospital, London, United Kingdom
- * E-mail:
| |
Collapse
|
42
|
Obar JJ, Hohl TM, Cramer RA. New advances in invasive aspergillosis immunobiology leading the way towards personalized therapeutic approaches. Cytokine 2016; 84:63-73. [PMID: 27253487 DOI: 10.1016/j.cyto.2016.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/16/2016] [Indexed: 01/07/2023]
Abstract
Invasive aspergillosis (IA) remains a devastating disease in immune compromised patients despite significant advances in our understanding of fungal virulence and host defense mechanisms. In this review, we summarize important research advances in the fight against IA with particular focus on early events in the interactions between Aspergillus fumigatus and the host that occur in the respiratory tract. Advances in understanding mechanisms of immune effector cell recruitment, antifungal effector mechanisms, and how the dynamic host-fungal interaction alters the local microenvironment to effect outcomes are highlighted. These advances illustrate exciting new therapeutic opportunities, but also emphasize the importance of understanding each unique fungus-host interaction for improving patient outcomes.
Collapse
Affiliation(s)
- Joshua J Obar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.
| | - Tobias M Hohl
- Infectious Disease Service, Department of Medicine, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY, United States; Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY, United States.
| | - Robert A Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.
| |
Collapse
|
43
|
Wang X, Zhang Y, Peng Y, Hutchinson MR, Rice KC, Yin H, Watkins LR. Pharmacological characterization of the opioid inactive isomers (+)-naltrexone and (+)-naloxone as antagonists of toll-like receptor 4. Br J Pharmacol 2016; 173:856-69. [PMID: 26603732 DOI: 10.1111/bph.13394] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 11/11/2015] [Accepted: 11/11/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The toll-like receptor TLR4 is involved in neuropathic pain and in drug reward and reinforcement. The opioid inactive isomers (+)-naltrexone and (+)-naloxone act as TLR4 antagonists, reversing neuropathic pain and reducing opioid and cocaine reward and reinforcement. However, how these agents modulate TLR4 signalling is not clear. Here, we have elucidated the molecular mechanism of (+)-naltrexone and (+)-naloxone on TLR4 signalling. EXPERIMENTAL APPROACH BV-2 mouse microglial cell line, primary rat microglia and primary rat peritoneal macrophages were treated with LPS and TLR4 signalling inhibitors. Effects were measured using Western blotting, luciferase reporter assays, fluorescence microscopy and ELISA KEY RESULTS: (+)-Naltrexone and (+)-naloxone were equi-potent inhibitors of the LPS-induced TLR4 downstream signalling and induction of the pro-inflammatory factors NO and TNF-α. Similarly, (+)-naltrexone or (+)-naloxone inhibited production of reactive oxygen species and increased microglial phagocytosis, induced by LPS. However, (+)-naltrexone and (+)-naloxone did not directly inhibit the increased production of IL-1β, induced by LPS. The drug interaction of (+)-naloxone and (+)-naltrexone was additive. (+)-Naltrexone or (+)-naloxone inhibited LPS-induced activation of IFN regulatory factor 3 and production of IFN-β. However, they did not inhibit TLR4 signalling via the activation of either NF-κB, p38 or JNK in these cellular models. CONCLUSIONS AND IMPLICATIONS (+)-Naltrexone and (+)-naloxone were TRIF-IFN regulatory factor 3 axis-biased TLR4 antagonists. They blocked TLR4 downstream signalling leading to NO, TNF-α and reactive oxygen species. This pattern may explain, at least in part, the in vivo therapeutic effects of (+)-naltrexone and (+)-naloxone.
Collapse
Affiliation(s)
- X Wang
- Chemical Biology Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Y Zhang
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - Y Peng
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - M R Hutchinson
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - K C Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20892, USA
| | - H Yin
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - L R Watkins
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, 80309, USA
| |
Collapse
|
44
|
Wójtowicz A, Lecompte TD, Bibert S, Manuel O, Rüeger S, Berger C, Boggian K, Cusini A, Garzoni C, Khanna N, Mueller NJ, Meylan PR, Pascual M, van Delden C, Bochud PY. Reply to Cunha et al. Clin Infect Dis 2015; 61:1894-5. [PMID: 26261206 DOI: 10.1093/cid/civ681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Thanh Doco Lecompte
- Service of Transplantation and Service of Infectious Diseases, University Hospitals of Geneva
| | - Stéphanie Bibert
- Infectious Diseases Service, University Hospital and University of Lausanne
| | - Oriol Manuel
- Infectious Diseases Service, University Hospital and University of Lausanne Transplantation Center, Department of Surgery
| | - Sina Rüeger
- Institute of Social and Preventive Medicine, University Hospital and University of Lausanne Swiss Institute of Bioinformatics, Lausanne
| | - Christoph Berger
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zürich
| | - Katia Boggian
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen
| | - Alexia Cusini
- Department of Infectious Diseases, Inselspital, Bern University Hospital and University of Bern
| | - Christian Garzoni
- Department of Infectious Diseases, Inselspital, Bern University Hospital and University of Bern Departments of Internal Medicine and Infectious Disease, Clinica Luganese, Lugano
| | - Nina Khanna
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Basel
| | - Nicolas J Mueller
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zurich
| | - Pascal R Meylan
- Infectious Diseases Service, University Hospital and University of Lausanne Institute of Microbiology, University Hospital and University of Lausanne, Switzerland
| | | | - Christian van Delden
- Service of Transplantation and Service of Infectious Diseases, University Hospitals of Geneva
| | - Pierre-Yves Bochud
- Infectious Diseases Service, University Hospital and University of Lausanne
| | | |
Collapse
|
45
|
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
|
46
|
Wójtowicz A, Lecompte TD, Bibert S, Manuel O, Rüeger S, Berger C, Boggian K, Cusini A, Garzoni C, Hirsch H, Khanna N, Mueller NJ, Meylan PR, Pascual M, van Delden C, Bochud PY. PTX3Polymorphisms and Invasive Mold Infections After Solid Organ Transplant: Figure 1. Clin Infect Dis 2015; 61:619-22. [DOI: 10.1093/cid/civ386] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/29/2015] [Indexed: 11/13/2022] Open
|
47
|
Oliveira-Coelho A, Rodrigues F, Campos A, Lacerda JF, Carvalho A, Cunha C. Paving the way for predictive diagnostics and personalized treatment of invasive aspergillosis. Front Microbiol 2015; 6:411. [PMID: 25999936 PMCID: PMC4419722 DOI: 10.3389/fmicb.2015.00411] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/20/2015] [Indexed: 01/24/2023] Open
Abstract
Invasive aspergillosis (IA) is a life-threatening fungal disease commonly diagnosed among individuals with immunological deficits, namely hematological patients undergoing chemotherapy or allogeneic hematopoietic stem cell transplantation. Vaccines are not available, and despite the improved diagnosis and antifungal therapy, the treatment of IA is associated with a poor outcome. Importantly, the risk of infection and its clinical outcome vary significantly even among patients with similar predisposing clinical factors and microbiological exposure. Recent insights into antifungal immunity have further highlighted the complexity of host-fungus interactions and the multiple pathogen-sensing systems activated to control infection. How to decode this information into clinical practice remains however, a challenging issue in medical mycology. Here, we address recent advances in our understanding of the host-fungus interaction and discuss the application of this knowledge in potential strategies with the aim of moving toward personalized diagnostics and treatment (theranostics) in immunocompromised patients. Ultimately, the integration of individual traits into a clinically applicable process to predict the risk and progression of disease, and the efficacy of antifungal prophylaxis and therapy, holds the promise of a pioneering innovation benefiting patients at risk of IA.
Collapse
Affiliation(s)
- Ana Oliveira-Coelho
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal ; ICVS/3B's - PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - Fernando Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal ; ICVS/3B's - PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - António Campos
- Serviço de Transplantação de Medula Óssea, Instituto Português de Oncologia do Porto , Porto, Portugal
| | - João F Lacerda
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa , Lisboa, Portugal ; Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria , Lisboa, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal ; ICVS/3B's - PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal ; ICVS/3B's - PT Government Associate Laboratory , Braga/Guimarães, Portugal
| |
Collapse
|