1
|
Chaves MM. Neutrophils and purinergic signaling: Partners in the crime against Leishmania parasites? Biochimie 2025; 232:43-53. [PMID: 39855456 DOI: 10.1016/j.biochi.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
The parasite of the genus Leishmania is the causative agent of diseases that affect humans called leishmaniasis. These diseases affect millions of people worldwide and the currently existing drugs are either very toxic or the parasites acquire resistance. Therefore, new elimination mechanisms need to be elucidated so that new therapeutic strategies can be developed. Much has already been discussed about the role of neutrophils in Leishmania infection, and their participation is still controversial. A recent study showed that receptors present in the neutrophil membrane, the purinergic receptors, can control the infection when activated, but the triggering mechanism has not been elucidated. In this review, we will address the possible participation of purinergic receptors expressed in the neutrophil extracellular membrane that may be participating in the detection of Leishmania infection and their possible effects during parasitism.
Collapse
Affiliation(s)
- Mariana M Chaves
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Bio-Manguinhos, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Fischer V, Ignatius A, Schmidt-Bleek K, Duda G, Haffner-Luntzer M. Using artificial intelligence-based software for an unbiased discrimination of immune cell subtypes in the fracture hematoma and bone marrow of non-osteoporotic and osteoporotic mice. PLoS One 2025; 20:e0322542. [PMID: 40299871 PMCID: PMC12040195 DOI: 10.1371/journal.pone.0322542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/24/2025] [Indexed: 05/01/2025] Open
Abstract
It is well established that the early inflammatory response following fracture is essential for initiating subsequent bone regeneration. An imbalance in inflammation, whether within the innate or adaptive immune response, can result in impaired fracture healing. In our previous studies, we demonstrated that, for example, mice with ovariectomy-induced osteoporosis exhibit altered immune cell populations in the early fracture hematoma and bone marrow, leading to delayed healing. These analyses were conducted using conventional FACS/flow cytometry software, where surface marker expression was assessed using a single threshold based on isotype controls-a binary "yes or no" decision. Recent advances have highlighted that immune cell populations are often more heterogeneous, with distinct phenotypic subgroups depending on their polarization status. This has been particularly well documented for macrophage subpopulations (M1, M2, and intermediate polarization states). In light of this, we employed a commercially available artificial intelligence-based clustering software (Cytolution) to more accurately and objectively identify immune cell subpopulations. We re-analyzed flow cytometry raw data from fracture hematoma and bone marrow of non-osteoporotic and osteoporotic mice at day 1 after fracture. Our findings revealed distinct subclusters for granulocytes (27 subclusters), macrophages (7 subclusters), B cells (4 subclusters), and T cells (6 subclusters) within the fracture hematoma and bone marrow. Comparing osteoporotic and non-osteoporotic mice, we observed an increased abundance of a specific B cell subpopulation in osteoporotic mice, alongside a significant reduction of a particular granulocyte subpopulation in the early fracture hematoma. Several subclusters of granulocytes, T cells, and macrophages were also altered in the bone marrow. The specific role of these immune cell subclusters remains to be investigated in the future. These results suggest that AI-based clustering may provide a powerful tool for identifying immune cell phenotypes during bone regeneration, offering a more nuanced understanding of flow cytometry data.
Collapse
Affiliation(s)
- Verena Fischer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | | | - Georg Duda
- Julius Wolff Institute, Berlin Institute of Health, Charité, Berlin, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
- Julius Wolff Institute, Berlin Institute of Health, Charité, Berlin, Germany
| |
Collapse
|
3
|
Viswanathan G, Hughes EJ, Gan M, Xet-Mull AM, Alexander G, Swain-Lenz D, Liu Q, Tobin DM. Granuloma Dual RNA-Seq Reveals Composite Transcriptional Programs Driven by Neutrophils and Necrosis within Tuberculous Granulomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.26.650783. [PMID: 40391323 PMCID: PMC12087985 DOI: 10.1101/2025.04.26.650783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Mycobacterial granulomas lie at the center of tuberculosis (TB) pathogenesis and represent a unique niche where infecting bacteria survive in nutrient-restricted conditions and in the face of a host immune response. The granuloma's necrotic core, where bacteria reside extracellularly in humans, is difficult to assess in many experimentally tractable models. Here, using necrotic mycobacterial granulomas in adult zebrafish, we develop dual RNA-seq across different host genotypes to identify the transcriptional alterations that enable bacteria to survive within this key microenvironment. Through pharmacological and genetic interventions, we find that neutrophils within mature, necrotic granulomas promote bacterial growth, in part through upregulation of the bacterial devR regulon. We identify conserved suites of bacterial transcriptional programs induced only in the context of this unique necrotic extracellular niche, including bacterial modules related to K + transport and rpf genes. Analysis of Mycobacterium tuberculosis strains across diverse lineages and human populations suggests that granuloma-specific transcriptional modules are targets for bacterial genetic adaptation in the context of human infection. Summary sentence Dual host-pathogen transcriptional profiling defines granuloma-specific programs during mycobacterial infection.
Collapse
|
4
|
Kanewska A, Lackner I, Friedrich A, Winkelmann M, Rojewski M, Weber B, Preßmar J, Perl M, Schrezenmeier H, Kalbitz M. Immunomodulatory and cardio-protective effects of differentially originated multipotent mesenchymal stroma cells during polymicrobial sepsis in mice. Eur J Trauma Emerg Surg 2025; 51:178. [PMID: 40253667 PMCID: PMC12009780 DOI: 10.1007/s00068-025-02862-2] [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: 12/27/2024] [Accepted: 04/03/2025] [Indexed: 04/22/2025]
Abstract
PURPOSE Sepsis is a life-threatening condition with cardiac complications being an independent predictor of poor outcome. Although their mechanisms have been widely investigated, therapeutic options remain limited. One promising therapeutic tool are mesenchymal stromal cells (MSCs). The aim of this study is to investigate the immunomodulatory effects of human MSCs from two different sources (bone marrow/BMMSC and adipose tissue/ASC) and to evaluate their cardioprotective potential. METHODS 60 adult male C57BL/6 mice were divided into sham, sepsis (cecal ligation puncture (CLP)) and two i.v. treatment groups CLP + human BMMSC and CLP + human ASC with 5 animals in each group. The observation periods were 8, 24 and 72 h. Left ventricular tissue was analyzed histologically, by qPCR (C3ar, C5ar1, Il-1b, Il-6, Il-10, Tlr2, Tlr4, Tnfa, and Nlrp3) and western blot. Cardiac damage markers troponin I and heart fatty acid binding protein (HFABP) were detected in serum by ELISA. RESULTS Troponin I and HFABP were significantly increased in CLP group after 8 h compared to sham. In cardiac tissue the expression of C3ar, C5ar1, Il-1b, Il-6, Il-10, Tlr2, Tlr4, Tnfa and Nlrp3 inflammasome was upregulated up to 24h after CLP compared to sham. After BMMSC treatment, C3ar as well as C5ar, Tlr2 and Il-10 mRNA expression in left ventricle was downregulated compared to CLP, whereas ASC treatment was associated with the downregulation of Il-6 and Nlrp3. CONCLUSIONS CLP-induced polymicrobial sepsis in mice was associated with cardiac damage and increased inflammation in left ventricular tissue. Therapeutic systemic application of human BMMSC and ASC ameliorated damage and inflammation in the heart.
Collapse
Affiliation(s)
- Anna Kanewska
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Ina Lackner
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Traumatology, Hand, Plastic and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Anne Friedrich
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Martina Winkelmann
- Institute for Transfusion Medicine, University Medical Center Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service, Ulm, Germany
| | - Markus Rojewski
- Institute for Transfusion Medicine, University Medical Center Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service, Ulm, Germany
| | - Birte Weber
- Department of Trauma Surgery and Orthopedics, University Hospital Frankfurt, Goethe-University, Frankfurt Am Main, Germany
| | - Jochen Preßmar
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Traumatology, Hand, Plastic and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Mario Perl
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University Medical Center Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service, Ulm, Germany
| | - Miriam Kalbitz
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Traumatology, Hand, Plastic and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
- Military Medical City Hospital (MMCH), Doha, Qatar
| |
Collapse
|
5
|
Jovanovski D, Wohlgemuth L, Lessing PML, Hüsken D, Koller AS, Thomaß BD, Müller P, Mannes M, Nungeß S, Jovanovska M, Mühling B, Liebold A, Huber-Lang M, Messerer DAC. Multimodal monitoring of neutrophil activity during cardiac surgery. Front Immunol 2025; 16:1504944. [PMID: 40151619 PMCID: PMC11947689 DOI: 10.3389/fimmu.2025.1504944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiac surgery and the associated ischemia-reperfusion injury trigger an inflammatory response, which, in turn, can contribute to organ damage, prolonged hospitalization, and mortality. Therefore, the present study performed comprehensive monitoring of neutrophil-related inflammation in patients who underwent aortic valve surgery, including extracorporeal circulation. Neutrophil-related inflammation, as well as alterations in cellular physiology, phenotype, and function, were analyzed by flow cytometry, ELISA, and microscopy. Neutrophil activation occurred intraoperatively and preceded the upregulation of conventional inflammatory markers such as C-reactive protein and interleukin-6. Perioperatively, neutrophils maintained a stable response to platelet-activating factor (PAF) with regard to CD11b and CD66b expression but showed a decreased response in CD10. Postoperatively, neutrophils exhibited marked alterations in PAF-induced depolarization, while reactive oxygen species generation and phagocytic activity remained largely stable. Surprisingly, platelet-neutrophil complex formation was severely impaired intraoperatively but returned to normal levels postoperatively. Further studies are needed to elucidate the implications of these intraoperative and postoperative changes in neutrophil and platelet activity with respect to a potential immune dysfunction that temporarily increases susceptibility to infectious or hemostatic complications.
Collapse
Affiliation(s)
- Darko Jovanovski
- Department of Cardiothoracic and Vascular Surgery, University Hospital Ulm, Ulm, Germany
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | | | - Dominik Hüsken
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | | | - Bertram Dietrich Thomaß
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Paul Müller
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Marco Mannes
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Sandra Nungeß
- Institute of Transfusion Medicine, University Hospital Ulm, Ulm, Germany
| | - Marta Jovanovska
- Department of Cardiothoracic and Vascular Surgery, University Hospital Ulm, Ulm, Germany
| | - Bernd Mühling
- Department of Cardiothoracic and Vascular Surgery, University Hospital Ulm, Ulm, Germany
| | - Andreas Liebold
- Department of Cardiothoracic and Vascular Surgery, University Hospital Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | | |
Collapse
|
6
|
Fernandes DC, Silva-de-França F, Pohl PC, Eto SF, Sardinha LR, Lambris JD, Tambourgi DV. Cp40-mediated complement C3 inhibition dampens inflammasome activation and inflammatory mediators storm induced by Bitis arietans venom. Int Immunopharmacol 2025; 147:113701. [PMID: 39809101 DOI: 10.1016/j.intimp.2024.113701] [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: 10/01/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
The complement system plays a crucial role in various pathophysiological conditions, including snake envenomation. In this study, we investigated the effects of Bitis arietans venom on the complement system using an ex vivo human whole blood model. Our findings demonstrate that B. arietans venom was able to activate the complement system, leading to a significant increase in the production of anaphylatoxins (C3a/C3a-desArg, C5a/C5a-desArg) and the soluble Terminal Complement Complex (sTCC). Inhibition of the C3 component by Cp40, a C3-C3b inhibitor, resulted in the reduction of C3a/C3a-desArg, C5a/C5a-desArg, and sTCC levels to baseline in venom-stimulated samples. Furthermore, treatment with Cp40 promoted a substantial decrease in the production of pro-inflammatory mediators, such as Prostaglandin E2 (PGE2), IL-8/CXCL8, MCP-1/CCL2, and MIG/CXCL9. To further elucidate the molecular mechanisms, we utilized the THP-1 cell line differentiated into M0 macrophages. Incubation of these macrophages with human plasma, from the human whole blood treated with B. arietans venom, resulted in the expression of the NLRP3 inflammasome and the production of IL-8 and IL-1β. Importantly, Cp40 was able to diminish the production of these cytokines, as well as the levels of ASC and caspase-1 proteins. In conclusion, our results indicate that the inhibition of the complement by Cp40 at C3/C3b level can modulate the inflammatory response and inflammasome activation induced by B. arietans venom. These findings suggest that complement inhibition may be a promising therapeutic approach for managing the inflammatory complications associated with this snake envenomation.
Collapse
Affiliation(s)
- Dayanne Carla Fernandes
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil
| | - Felipe Silva-de-França
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil
| | | | | | | | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Denise V Tambourgi
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil.
| |
Collapse
|
7
|
Vygonskaya M, Wu Y, Price TJ, Chen Z, Smith MT, Klyne DM, Han FY. The role and treatment potential of the complement pathway in chronic pain. THE JOURNAL OF PAIN 2025; 27:104689. [PMID: 39362355 DOI: 10.1016/j.jpain.2024.104689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
The role of the complement system in pain syndromes has garnered attention on the back of preclinical and clinical evidence supporting its potential as a target for new analgesic pharmacotherapies. Of the components that make up the complement system, component 5a (C5a) and component 3a (C3a) are most strongly and consistently associated with pain. Receptors for C5a are widely found in immune resident cells (microglia, astrocytes, sensory neuron-associated macrophages (sNAMs)) in the central nervous system (CNS) as well as hematogenous immune cells (mast cells, macrophages, T-lymphocytes, etc.). When active, as is often observed in chronic pain conditions, these cells produce various inflammatory mediators including pro-inflammatory cytokines. These events can trigger nervous tissue inflammation (neuroinflammation) which coexists with and potentially maintains peripheral and central sensitization. C5a has a likely critical role in initiating this process highlighting its potential as a promising non-opioid target for treating pain. This review summarizes the most up-to-date research on the role of the complement system in pain with emphasis on the C5 pathway in peripheral tissue, dorsal root ganglia (DRG) and the CNS, and explores advances in complement-targeted drug development and sex differences. A perspective on the optimal application of different C5a inhibitors for different types (e.g., neuropathic, post-surgical and chemotherapy-induced pain, osteoarthritis pain) and stages (e.g., acute, subacute, chronic) of pain is also provided to help guide future clinical trials. PERSPECTIVE: This review highlights the role and mechanisms of complement components and their receptors in physiological and pathological pain. The potential of complement-targeted therapeutics for the treatment of chronic pain is also explored with a focus on C5a inhibitors to help guide future clinical trials.
Collapse
Affiliation(s)
- Marina Vygonskaya
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Youzhi Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Theodore J Price
- Center for Advanced Pain Studies, Department of Neuroscience, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Zhuo Chen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Maree T Smith
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - David M Klyne
- NHMRC Centre of Clinical Research Excellence in Spinal Pain, Injury and Health, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Felicity Y Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
8
|
Bakker NAM, Burrello C, de Visser KE. Ex vivo assessment of human neutrophil motility and migration. Methods Cell Biol 2024; 191:115-133. [PMID: 39824552 DOI: 10.1016/bs.mcb.2024.10.008] [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] [Indexed: 01/20/2025]
Abstract
Neutrophils are pivotal in orchestrating tumor-induced systemic inflammation and are increasingly recognized for their critical involvement in both the initiation and progression of cancer. A fundamental facet of neutrophil biology is their migratory capacity, which enables them to extravasate and infiltrate tumors in other tissues, where they carry out essential effector functions. Unraveling the intricate mechanisms of neutrophil motility and migration is crucial for comprehending immune responses and inflammatory processes, shedding light on their substantial contribution to cancer progression. Here, we provide a comprehensive protocol to assess direct ex vivo motility and migration of freshly isolated human neutrophils, offering valuable insights into their behavior.
Collapse
Affiliation(s)
- Noor A M Bakker
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Claudia Burrello
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Karin E de Visser
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands; Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
9
|
Warner S, Teague HL, Ramos-Benitez MJ, Panicker S, Allen K, Gairhe S, Moyer T, Parachalil Gopalan B, Douagi I, Shet A, Kanthi Y, Suffredini AF, Chertow DS, Strich JR. R406 reduces lipopolysaccharide-induced neutrophil activation. Cell Immunol 2024; 403-404:104860. [PMID: 39084187 PMCID: PMC11387147 DOI: 10.1016/j.cellimm.2024.104860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Modulating SYK has been demonstrated to have impacts on pathogenic neutrophil responses in COVID-19. During sepsis, neutrophils are vital in early bacterial clearance but also contribute to the dysregulated immune response and organ injury when hyperactivated. Here, we evaluated the impact of R406, the active metabolite of fostamatinib, on neutrophils stimulated by LPS. We demonstrate that R406 was able to effectively inhibit NETosis, degranulation, ROS generation, neutrophil adhesion, and the formation of CD16low neutrophils that have been linked to detrimental outcomes in severe sepsis. Further, the neutrophils remain metabolically active, capable of releasing cytokines, perform phagocytosis, and migrate in response to IL-8. Taken together, this data provides evidence of the potential efficacy of utilizing fostamatinib in bacterial sepsis.
Collapse
Affiliation(s)
- Seth Warner
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Heather L Teague
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Marcos J Ramos-Benitez
- Basic Science Department, Microbiology Division, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Sumith Panicker
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kiana Allen
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Salina Gairhe
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Tom Moyer
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bindu Parachalil Gopalan
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Iyadh Douagi
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; NIH Center for Human Immunology, Inflammation, and Autoimmunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arun Shet
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anthony F Suffredini
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S Chertow
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA; Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey R Strich
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
10
|
Abstract
While the involvement of actin polymerization in cell migration is well-established, much less is known about the role of transmembrane water flow in cell motility. Here, we investigate the role of water influx in a prototypical migrating cell, the neutrophil, which undergoes rapid, directed movement to sites of injury, and infection. Chemoattractant exposure both increases cell volume and potentiates migration, but the causal link between these processes are not known. We combine single-cell volume measurements and a genome-wide CRISPR screen to identify the regulators of chemoattractant-induced neutrophil swelling, including NHE1, AE2, PI3K-gamma, and CA2. Through NHE1 inhibition in primary human neutrophils, we show that cell swelling is both necessary and sufficient for the potentiation of migration following chemoattractant stimulation. Our data demonstrate that chemoattractant-driven cell swelling complements cytoskeletal rearrangements to enhance migration speed.
Collapse
Affiliation(s)
- Tamas L Nagy
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Evelyn Strickland
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Orion D Weiner
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
11
|
Urbanska M, Guck J. Single-Cell Mechanics: Structural Determinants and Functional Relevance. Annu Rev Biophys 2024; 53:367-395. [PMID: 38382116 DOI: 10.1146/annurev-biophys-030822-030629] [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] [Indexed: 02/23/2024]
Abstract
The mechanical phenotype of a cell determines its ability to deform under force and is therefore relevant to cellular functions that require changes in cell shape, such as migration or circulation through the microvasculature. On the practical level, the mechanical phenotype can be used as a global readout of the cell's functional state, a marker for disease diagnostics, or an input for tissue modeling. We focus our review on the current knowledge of structural components that contribute to the determination of the cellular mechanical properties and highlight the physiological processes in which the mechanical phenotype of the cells is of critical relevance. The ongoing efforts to understand how to efficiently measure and control the mechanical properties of cells will define the progress in the field and drive mechanical phenotyping toward clinical applications.
Collapse
Affiliation(s)
- Marta Urbanska
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
12
|
Nagy TL, Strickland E, Weiner OD. Neutrophils actively swell to potentiate rapid migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.15.540704. [PMID: 37292824 PMCID: PMC10245588 DOI: 10.1101/2023.05.15.540704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
While the involvement of actin polymerization in cell migration is well-established, much less is known about the role of transmembrane water flow in cell motility. Here, we investigate the role of water influx in a prototypical migrating cell, the neutrophil, which undergoes rapid, directed movement to sites of injury and infection. Chemoattractant exposure both increases cell volume and potentiates migration, but the causal link between these processes is not known. We combine single cell volume measurements and a genome-wide CRISPR screen to identify the regulators of chemoattractant-induced neutrophil swelling, including NHE1, AE2, PI3K-gamma, and CA2. Through NHE1 inhibition in primary human neutrophils, we show that cell swelling is both necessary and sufficient for the potentiation of migration following chemoattractant stimulation. Our data demonstrate that chemoattractant-driven cell swelling complements cytoskeletal rearrangements to enhance migration speed.
Collapse
Affiliation(s)
- Tamas L Nagy
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Evelyn Strickland
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Orion D Weiner
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Shen B, Shen Q, Zeng Q, Zhang L, Li X. Silenced-C5ar1 improved multiple organ injury in sepsis rats via inhibiting neutrophil extracellular trap. J Mol Histol 2024; 55:69-81. [PMID: 38165570 PMCID: PMC10830609 DOI: 10.1007/s10735-023-10172-3] [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: 03/16/2023] [Accepted: 10/21/2023] [Indexed: 01/04/2024]
Abstract
Sepsis has a systemic inflammatory response syndrome caused by infection. While neutrophils play contradictory roles in different stages of sepsis. Neutrophils have been proven to play an antibacterial role by producing neutrophil extracellular traps (NETs). Although the NET is beneficial to bacteria resistance, abnormal NET increases tissue damage. The complement C5a receptor 1 (C5ar1) is a gene related to strong inflammatory reactions and is found to be associated with inflammatory factors. This study found that there were 45 down-regulated genes and 704 up-regulated genes in sepsis rats by transcriptome sequencing. And those genes were significantly related to inflammation and immunity by GO and KEGG enrichment analysis involving the chemokine signaling pathway, the Toll-like receptor (TLR) signaling pathway, and the Fc gamma R-mediated phagocytosis. Additionally, the C5ar1 gene was significantly upregulated with interesting potential in sepsis and used for further study. This study used cecum ligation and puncture (CLP) rats that were respectively injected intravenously with PBS or the lentivirus vector to explore the effect of C5ar1 on CLP rats. It demonstrated that silenced- C5ar1 inhibited the ALT, AST, BUN, and CREA levels, improved the lung and spleen injury, and reduced the TNF-α, IL-6, IL-1β, IL-10, cf-DNA, and cfDNA/MPO levels. Additionally, silenced C5ar1 inhibited the TLR2, TLR4, and peptidylarginine deiminase 4 expression levels, which suggested the improvement of silenced C5ar1 on sepsis via inhibiting NETs and the TLR signaling pathway. This study provides a basis and new direction for the study of treatment on sepsis.
Collapse
Affiliation(s)
- Bin Shen
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, 313000, China
| | - Qikai Shen
- Department of Intensive Care Units, Huzhou Central Hospital, Huzhou, 313000, China
| | - Qingqiu Zeng
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, 313000, China
| | - Lingyan Zhang
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, 313000, China
| | - Xiaofeng Li
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, 313000, China.
| |
Collapse
|
14
|
Fu X, Iglesias-Álvarez D, García-Campos A, Martínez-Monzonís MA, Almenglo C, Martinez-Cereijo JM, Reija L, Fernandez ÁL, Gonzalez-Juanatey JR, Rodriguez-Manero M, Eiras S. Enhanced Levels of Adiposity, Stretch and Fibrosis Markers in Patients with Coexistent Heart Failure and Atrial Fibrillation. J Cardiovasc Transl Res 2024; 17:13-23. [PMID: 37878196 DOI: 10.1007/s12265-023-10454-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/11/2023] [Indexed: 10/26/2023]
Abstract
The coexistence of heart failure (HF) and atrial fibrillation (AF) worsens the prognosis of patients. We aimed to study the inflammation, metabolism, adiposity, and fibrosis markers on epicardial and subcutaneous fat and blood, and their relationship with HF and AF. Samples from 185 patients undergoing cardiac surgery were collected. Levels of multi-markers on fat biopsies and plasma were analyzed. Patients were grouped by HF or AF presence. Plasma adiposity markers were increased in AF patients, while increased stretch markers correlated with HF. Patients with both AF and HF had higher ANP and GDF-15 levels. After excluding AF patients, plasma FABP4 was identified as the main HF predictor. Fat biopsies from AF patients showed an enhanced inflammatory profile. Higher levels of adiposity markers are associated with AF or HF, and higher stretch and fibrosis markers with combined AF and HF, suggesting a role of adiposity-fibrosis pathway in HF and AF coexistence.
Collapse
Affiliation(s)
- Xiaoran Fu
- Translational Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Diego Iglesias-Álvarez
- Cardiovascular Area, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana García-Campos
- Cardiovascular Area, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- CIBERCV, Madrid, Spain
| | | | - Cristina Almenglo
- Translational Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Laura Reija
- Heart Surgery Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ángel Luis Fernandez
- CIBERCV, Madrid, Spain
- Heart Surgery Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Jose Ramón Gonzalez-Juanatey
- Cardiovascular Area, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- CIBERCV, Madrid, Spain
- Medicine Department, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Moises Rodriguez-Manero
- Translational Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- Cardiovascular Area, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- CIBERCV, Madrid, Spain
| | - Sonia Eiras
- Translational Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.
- CIBERCV, Madrid, Spain.
| |
Collapse
|
15
|
Schmidt C, Weißmüller S, Heinz CC. Multifaceted Tissue-Protective Functions of Polyvalent Immunoglobulin Preparations in Severe Infections-Interactions with Neutrophils, Complement, and Coagulation Pathways. Biomedicines 2023; 11:3022. [PMID: 38002022 PMCID: PMC10669904 DOI: 10.3390/biomedicines11113022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/30/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Severe infections induce immune defense mechanisms and initial tissue damage, which produce an inflammatory neutrophil response. Upon dysregulation of these responses, inflammation, further tissue damage, and systemic spread of the pathogen may occur. Subsequent vascular inflammation and activation of coagulation processes may cause microvascular obstruction at sites distal to the primary site of infection. Low immunoglobulin (Ig) M and IgG levels have been detected in patients with severe infections like sCAP and sepsis, associated with increased severity and mortality. Based on Ig's modes of action, supplementation with polyvalent intravenous Ig preparations (standard IVIg or IgM/IgA-enriched Ig preparations) has long been discussed as a treatment option for severe infections. A prerequisite seems to be the timely administration of Ig preparations before excessive tissue damage has occurred and coagulopathy has developed. This review focuses on nonclinical and clinical studies that evaluated tissue-protective activities resulting from interactions of Igs with neutrophils, complement, and the coagulation system. The data indicate that coagulopathy, organ failure, and even death of patients can possibly be prevented by the timely combined interactions of (natural) IgM, IgA, and IgG with neutrophils and complement.
Collapse
Affiliation(s)
- Carolin Schmidt
- Department of Corporate Clinical Research and Development, Biotest AG, 63303 Dreieich, Germany
| | | | - Corina C Heinz
- Department of Corporate Clinical Research and Development, Biotest AG, 63303 Dreieich, Germany
| |
Collapse
|
16
|
Kwantwi LB. Overcoming anti-PD-1/PD-L1 immune checkpoint blockade resistance: the role of macrophage, neutrophils and mast cells in the tumor microenvironment. Clin Exp Med 2023; 23:3077-3091. [PMID: 37022584 DOI: 10.1007/s10238-023-01059-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023]
Abstract
In recent years, the anti-PD-1/PD-L1 blockade has become a game changer in cancer treatment following the unprecedented response rate. Regardless of the substantial therapy efficacy across various cancer types, some patients do not still respond to these therapies, indicating that a deeper understanding of the mechanisms of anti-PD-1/PD-L1 resistance is highly important. To overcome such resistance, the tumor-induced immunosuppressive mechanisms have been focused and several suppressor cell populations in the tumor microenvironment have been identified. Among these cells, macrophages, neutrophils, and mast cells are known to play key roles in anti-PD-1/PD-L1 resistance. Hence, gaining control over these innate immune cells can open opportunities for breaking tumor resistance to immune checkpoint inhibitors. Herein, a summary of the role of macrophages, neutrophils, and mast cells in anti-PD-1/PD-L1 resistance has been described. Also, strategies to overcome their therapeutic resistance to anti-PD-1/PD-L1 have been discussed.
Collapse
Affiliation(s)
- Louis Boafo Kwantwi
- Department of Medical Imaging Sciences, Klintaps College of Health and Allied Sciences, Accra, DTD. TDC, 30A Klagon, Com. 19, Tema, Ghana.
| |
Collapse
|
17
|
Kuhn A, Riegger J, Teixeira GQ, Huber-Lang M, Lambris JD, Neidlinger-Wilke C, Brenner RE. Terminal Complement Activation Is Induced by Factors Released from Endplate Tissue of Disc Degeneration Patients and Stimulates Expression of Catabolic Enzymes in Annulus Fibrosus Cells. Cells 2023; 12:cells12060887. [PMID: 36980228 PMCID: PMC10047197 DOI: 10.3390/cells12060887] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Terminal complement complex (TCC) deposition was identified in human degenerated discs. To clarify the role of terminal complement activation in disc degeneration (DD), we investigated respective activating mechanisms and cellular effects in annulus fibrosus (AF) cells. Isolated cells from human AF, nucleus pulposus (NP), and endplate (EP) were stimulated with human serum alone or with zymosan and treated with either the C3 inhibitor Cp40 or the C5 antibody eculizumab. Complement activation was determined via anaphylatoxin generation and TCC deposition detection. Thereby, induced catabolic effects were evaluated in cultured AF cells. Moreover, C5 cleavage under degenerative conditions in the presence of AF cells was assessed. Zymosan-induced anaphylatoxin generation and TCC deposition was significantly suppressed by both complement inhibitors. Zymosan induced gene expression of ADAMTS4, MMP1, and COX2. Whereas the C3 blockade attenuated the expression of ADAMTS4, the C5 blockade reduced the expression of ADAMTS4, MMP1, and COX2. Direct C5 cleavage was significantly enhanced by EP conditioned medium from DD patients and CTSD. These results indicate that terminal complement activation might be functionally involved in the progression of DD. Moreover, we found evidence that soluble factors secreted by degenerated EP tissue can mediate direct C5 cleavage, thereby contributing to complement activation in degenerated discs.
Collapse
Affiliation(s)
- Amelie Kuhn
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University, 89081 Ulm, Germany
| | - Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University, 89081 Ulm, Germany
| | - Graciosa Q. Teixeira
- Institute of Orthopedic Research and Biomechanics, Trauma Research Centre, Ulm University, 89081 Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cornelia Neidlinger-Wilke
- Institute of Orthopedic Research and Biomechanics, Trauma Research Centre, Ulm University, 89081 Ulm, Germany
| | - Rolf E. Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University, 89081 Ulm, Germany
- Correspondence: ; Tel.: +49-(0)731-500-63280
| |
Collapse
|
18
|
Kempter E, Amoroso M, Kupfer S, Lupu L, Kustermann M, Scheurer J, Baumann B, Wirth T, Gündel H, Straub RH, Strauß G, Huber-Lang M, Langgartner D, Reber SO. The PMN-MDSC - A key player in glucocorticoid resistance following combined physical and psychosocial trauma. Brain Behav Immun 2023; 108:148-161. [PMID: 36427809 DOI: 10.1016/j.bbi.2022.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/21/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
Stress-associated somatic and psychiatric disorders are often linked to non-resolving low-grade inflammation, which is promoted at least in part by glucocorticoid (GC) resistance of distinct immune cell subpopulations. While the monocyte/macrophage compartment was in the focus of many clinical and preclinical studies, the role of myeloid-derived suppressor cells (MDSCs) in stress-associated pathologies and GC resistance is less understood. As GC resistance is a clear risk factor for posttraumatic complications in patients on intensive care, the exact interplay of physical and psychosocial traumatization in the development of GC resistance needs to be further clarified. In the current study we employ the chronic subordinate colony housing (CSC) paradigm, a well-characterized mouse model of chronic psychosocial stress, to study the role of myeloid cells, in particular of MDSCs, in innate immune activation and GC resistance following combined psychosocial and physical (e.g., bite wounds) trauma. Our findings support the hypothesis that stress-induced neutrophils, polymorphonuclear (PMN)-MDSCs and monocytes/monocyte-like (MO)-MDSCs get primed and activated locally in the bone marrow as determined by toll-like receptor (TLR)2 upregulation and increased basal and lipopolysaccharide (LPS)-induced in vitro cell viability. These primed and activated myeloid cells emigrate into the peripheral circulation and subsequently, if CSC is accompanied by significant bite wounding, accumulate in the spleen. Here, PMN-MDSCs and monocytes/MO-MDSCs upregulate TLR4 expression, which exclusively in PMN-MDSCs promotes NF-κB hyperactivation upon LPS-stimulation, thereby exceeding the anti-inflammatory capacities of GCs and resulting in GC resistance.
Collapse
Affiliation(s)
- Elena Kempter
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Mattia Amoroso
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Sandra Kupfer
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Ludmila Lupu
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Monika Kustermann
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Jasmin Scheurer
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Harald Gündel
- Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Rainer H Straub
- Laboratory of Experimental Rheumatology and Neuroendocrine Immunology, Division of Rheumatology, Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Gudrun Strauß
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
19
|
Modulation of Neutrophil Activity by Soluble Complement Cleavage Products—An In-Depth Analysis. Cells 2022; 11:cells11203297. [PMID: 36291163 PMCID: PMC9600402 DOI: 10.3390/cells11203297] [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: 09/15/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/16/2022] Open
Abstract
The cellular and fluid phase-innate immune responses of many diseases predominantly involve activated neutrophil granulocytes and complement factors. However, a comparative systematic analysis of the early impact of key soluble complement cleavage products, including anaphylatoxins, on neutrophil granulocyte function is lacking. Neutrophil activity was monitored by flow cytometry regarding cellular (electro-)physiology, cellular activity, and changes in the surface expression of activation markers. The study revealed no major effects induced by C3a or C4a on neutrophil functions. By contrast, exposure to C5a or C5a des-Arg stimulated neutrophil activity as reflected in changes in membrane potential, intracellular pH, glucose uptake, and cellular size. Similarly, C5a and C5a des-Arg but no other monitored complement cleavage product enhanced phagocytosis and reactive oxygen species generation. C5a and C5a des-Arg also altered the neutrophil surface expression of several complement receptors and neutrophil activation markers, including C5aR1, CD62L, CD10, and CD11b, among others. In addition, a detailed characterization of the C5a-induced effects was performed with a time resolution of seconds. The multiparametric response of neutrophils was further analyzed by a principal component analysis, revealing CD11b, CD10, and CD16 to be key surrogates of the C5a-induced effects. Overall, we provide a comprehensive insight into the very early interactions of neutrophil granulocytes with activated complement split products and the resulting neutrophil activity. The results provide a basis for a better and, importantly, time-resolved and multiparametric understanding of neutrophil-related (patho-)physiologies.
Collapse
|
20
|
Paloschi MV, Boeno CN, Lopes JA, Rego CMA, Silva MDS, Santana HM, Serrath SN, Ikenohuchi YJ, Farias BJC, Felipin KP, Nery NM, Dos Reis VP, de Lima Lemos CT, Evangelista JR, da Silva Setúbal S, Soares AM, Zuliani JP. Reactive oxygen species-dependent-NLRP3 inflammasome activation in human neutrophils induced by l-amino acid oxidase derived from Calloselasma rhodostoma venom. Life Sci 2022; 308:120962. [PMID: 36113732 DOI: 10.1016/j.lfs.2022.120962] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/31/2022] [Accepted: 09/10/2022] [Indexed: 11/20/2022]
Abstract
l-Amino acid oxidase isolated from Calloselasma rhodostoma (Cr-LAAO) snake venom is a potent stimulus for neutrophil activation and production of inflammatory mediators, contributing to local inflammatory effects in victims of envenoming. Cr-LAAO triggered the activation of nicotinamide adenine dinucleotide phosphatase (NADPH) oxidase complex and protein kinase C (PKC)-α signaling protein for reactive oxygen species (ROS) production. This study aims to evaluate the ROS participation in the NLRP3 inflammasome complex activation in human neutrophil. Human neutrophils were isolated and stimulated for 1 or 2 h with RPMI (negative control), LPS (1 μg/mL, positive control) or Cr-LAAO (50 μg/mL). The neutrophil transcriptome was examined using the microarray technique, and RT-qPCR for confirmation of gene expression. Immunofluorescence assays for NLRP3, caspase-1, IL-1β and GSDMD proteins was performed by Western blot in the presence and/or absence of Apocynin, an inhibitor of NADPH oxidase. IL-1β release was also detected in the presence and/or absence of NLRP3, caspase-1 and NADPH oxidase inhibitors. Results showed that Cr-LAAO upregulated the expression of genes that participate in the NADPH oxidase complex formation and inflammasome assembly. NLRP3 was activated and accumulated in the cytosol forming punctas, indicating its activation. Gasdermin D was not cleaved but lactate dehydrogenase was released. Furthermore, ROS inhibition decreased the expression of NLRP3 inflammasome complex proteins, as observed by protein expression in the presence and/or absence of apocynin, an NADPH oxidase inhibitor. IL-1β was also released, and pharmacological inhibition of NLRP3, caspase-1, and ROS reduced the amount of released cytokine. This is the first report demonstrating the activation of the NLRP3 inflammasome complex via ROS generation by Cr-LAAO, which may lead to the development of local inflammatory effects observed in snakebite victims.
Collapse
Affiliation(s)
- Mauro Valentino Paloschi
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Charles Nunes Boeno
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Jéssica Amaral Lopes
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Cristina Matiele Alves Rego
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Milena Daniela Souza Silva
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Hallison Mota Santana
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Suzanne Nery Serrath
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Yoda Janaina Ikenohuchi
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Braz Junior Campos Farias
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Kátia Paula Felipin
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Neriane Monteiro Nery
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Valdison Pereira Dos Reis
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Caleb Torres de Lima Lemos
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Jaina Rodrigues Evangelista
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Sulamita da Silva Setúbal
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Andreimar Martins Soares
- Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia e Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Juliana Pavan Zuliani
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia e Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil.
| |
Collapse
|
21
|
Yu FF, Yuan Y, Ao Y, Hua L, Wang W, Cao Y, Xi J, Luan Y, Hou S, Zhang XY. A New Product of Bilirubin Degradation by H 2O 2 and Its Formation in Activated Neutrophils and in an Inflammatory Mouse Model. Biomolecules 2022; 12:biom12091237. [PMID: 36139076 PMCID: PMC9496627 DOI: 10.3390/biom12091237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Bilirubin (BR) is a tetrapyrrolic compound stemming from heme catabolism with diverse physiological functions. It can be oxidized by H2O2 to form several degradation products, some of which have been detected in vivo and may contribute to the pathogenesis of certain diseases. However, the oxidative degradation of BR is complex and the conditions that BR degradation occurs pathophysiologically remain obscure. Neutrophils are known to generate large amounts of reactive oxygen species, including H2O2, upon activation and they are mobilized to inflammatory sites; therefore, we hypothesized that activated neutrophils could cause BR degradation, which could occur at inflammatory sites. In the present study, we investigated BR degradation by H2O2 and identified hematinic acid (BHP1) and a new product BHP2, whose structure was characterized as 2,5-diformyl-4-methyl-1H-pyrrole-3-propanoic acid. An LC-MS/MS method for the quantitation of the two compounds was then established. Using the LC-MS/MS method, we observed the concentration-dependent formation of BHP1 and BHP2 in mouse neutrophils incubated with 10 and 30 μM of BR with the yields being 16 ± 3.2 and 31 ± 5.9 pmol/106 cells for BHP1, and 25 ± 4.4 and 71 ± 26 pmol/106 cells for BHP2, respectively. After adding phorbol 12-myristate 13-acetate, a neutrophil agonist, to 30 μM of BR-treated cells, the BHP1 yield increased to 43 ± 6.6 pmol/106 cells, whereas the BHP2 one decreased to 47 ± 9.2 pmol/106 cells. The two products were also detected in hemorrhagic skins of mice with dermal inflammation and hemorrhage at levels of 4.5 ± 1.9 and 0.18 ± 0.10 nmol/g tissue, respectively, which were significantly higher than those in the non-hemorrhagic skins. BHP2 was neurotoxic starting at 0.10 μM but BHP1 was not, as assessed using Caenorhabditis elegans as the animal model. Neutrophil-mediated BR degradation may be a universally pathophysiological process in inflammation and can be particularly important under pathological conditions concerning hemorrhage.
Collapse
Affiliation(s)
- Fei-Fei Yu
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yao Yuan
- Shanghai Jiao Tong University-Hangzhou Future Sci-Tech City Joint Research Center for Tumor Immunotherapy, Hangzhou Innovation Institute for Systems Oncology, Hangzhou 311121, China
| | - Yan Ao
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Li Hua
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wu Wang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
- Correspondence: (W.W.); (S.H.); (X.-Y.Z.)
| | - Yiyi Cao
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Xi
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yang Luan
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shangwei Hou
- Shanghai Jiao Tong University-Hangzhou Future Sci-Tech City Joint Research Center for Tumor Immunotherapy, Hangzhou Innovation Institute for Systems Oncology, Hangzhou 311121, China
- Correspondence: (W.W.); (S.H.); (X.-Y.Z.)
| | - Xin-Yu Zhang
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (W.W.); (S.H.); (X.-Y.Z.)
| |
Collapse
|
22
|
Neutrophils activated by membrane attack complexes increase the permeability of melanoma blood vessels. Proc Natl Acad Sci U S A 2022; 119:e2122716119. [PMID: 35960843 PMCID: PMC9388087 DOI: 10.1073/pnas.2122716119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cancer cell dissemination is the seed for metastasis and adversely linked to patients’ benefit. Critical for hematogenous dissemination is the entrance of the cancer cell into the circulation, which is regulated by vascular permeability within the primary tumor. Here, we describe pathophysiological communication between endothelial cells, tumor infiltrating neutrophils, and the complement system, with implications for vascular barrier opening and melanoma cell dissemination. Experiments in complement-deficient animals indicate that interference with complement-mediated activation of neutrophils stabilizes blood vessel integrity and abolishes the systemic spread of melanoma cells. The microenvironment of malignant melanomas defines the properties of tumor blood vessels and regulates infiltration and vascular dissemination of immune and cancer cells, respectively. Previous research in other cancer entities suggested the complement system as an essential part of the tumor microenvironment. Here, we confirm activation of the complement system in samples of melanoma patients and murine melanomas. We identified the tumor endothelium as the starting point of the complement cascade. Generation of complement-derived C5a promoted the recruitment of neutrophils. Upon contact with the vascular endothelium, neutrophils were further activated by complement membrane attack complexes (MACs). MAC-activated neutrophils release neutrophil extracellular traps (NETs). Close to the blood vessel wall, NETs opened the endothelial barrier as indicated by an enhanced vascular leakage. This facilitated the entrance of melanoma cells into the circulation and their systemic spread. Depletion of neutrophils or lack of MAC formation in complement component 6 (C6)–deficient animals protected the vascular endothelium and prevented vascular intravasation of melanoma cells. Our data suggest that inhibition of MAC-mediated neutrophil activation is a potent strategy to abolish hematogenous dissemination in melanoma.
Collapse
|
23
|
Chitosan and its derivatives as polymeric anti-viral therapeutics and potential anti-SARS-CoV-2 nanomedicine. Carbohydr Polym 2022; 290:119500. [PMID: 35550778 PMCID: PMC9020865 DOI: 10.1016/j.carbpol.2022.119500] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 01/07/2023]
Abstract
The coronavirus pandemic, COVID-19 has a global impact on the lives and livelihoods of people. It is characterized by a widespread infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), where infected patients may develop serious medical complications or even face death. Development of therapeutic is essential to reduce the morbidity and mortality of infected patients. Chitosan is a versatile biomaterial in nanomedicine and exhibits anti-microbial, anti-cancer and immunomodulatory properties. This review highlights the progress in chitosan design and application pertaining to the anti-viral effects of chitosan and chitosan derivatives (hydroxypropyl trimethylammonium, sulfate, carboxymethyl, bromine, sialylglycopolymer, peptide and phosphonium conjugates) as a function of molecular weight, degree of deacetylation, type of substituents and their degree and site of substitution. The physicochemical attributes of these polymeric therapeutics are identified against the possibility of processing them into nanomedicine which can confer a higher level of anti-viral efficacy. The designs of chitosan for the purpose of targeting SARS-CoV-2, as well as the ever-evolving strains of viruses with a broad spectrum anti-viral activity to meet pandemic preparedness at the early stages of outbreak are discussed.
Collapse
|
24
|
Almengló C, Fu X, Flores‐Arias MT, Fernández ÁL, Viñuela JE, Martínez‐Cereijo JM, Durán D, Rodríguez‐Mañero M, González‐Juanatey JR, Eiras S. Synergism between obesity and HFpEF on neutrophils phenotype and its regulation by adipose tissue-molecules and SGLT2i dapagliflozin. J Cell Mol Med 2022; 26:4416-4427. [PMID: 35818731 PMCID: PMC9357605 DOI: 10.1111/jcmm.17466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/30/2022] [Accepted: 06/15/2022] [Indexed: 11/27/2022] Open
Abstract
The adiposity invokes innate immune activity, coronary microvascular dysfunction and consequently heart failure preserved ejection fraction (HFpEF). Our aim was to study the neutrophils profile on obesity and cardiovascular disease and its regulation by adipose tissue-secretome and dapagliflozin. We have isolated neutrophils from patients undergoing open heart surgery (19 women and 51 men). Its migration activity was performed with culture-transwell, transcriptional studies of proteolytic enzymes, adhesion molecules or receptors were analysed by real-time PCR and proteomics (from 20 patients) analysis by TripleTOF mass spectrometer. Differentiated HL-60 (dHL-60) was used as a preclinical model on microfluidic for endothelial cells attaching assays and genes regulation with epicardial and subcutaneous fat secretomes from patients (3 women and 9 men) or dapagliflozin 1-10 μM treatments. The transcriptional and proteomics studies have determined higher levels of adhesion molecules in neutrophils from patients with obesity. The adhesion molecule CD11b levels were higher in those patients with the combined obesity and HFpEF factors (1.70 ± 0.06 a.u. without obesity, 1.72 ± 0.04 a.u. obesity or HFpEF without obesity and 1.79 ± 0.08 a.u. obesity and HFpEF; p < .01). While fat-secretome induces its upregulation, dapagliflozin can modulated it. Because CD11b upregulation is associated with higher neutrophils migration and adhesion into endothelial cells, dapagliflozin might modulate this mechanism on patients with obesity and HFpEF.
Collapse
Affiliation(s)
- Cristina Almengló
- Cardiology GroupHealth Research InstituteSantiago de CompostelaSpain
| | - Xiaoran Fu
- Translational Cardiology GroupHealth Research InstituteSantiago de CompostelaSpain
| | - María Teresa Flores‐Arias
- Photonics4 Life Research Group, Applied Physics Department, Facultade de Física and Facultade de Óptica e OptometríaUniversidade de Santiago de CompostelaSantiago de CompostelaSpain
| | - Ángel L. Fernández
- Heart Surgery DepartmentUniversity Hospital of Santiago de CompostelaSantiago de CompostelaSpain
- CIBERCV MadridMadridSpain
| | - Juan E. Viñuela
- Translational Cardiology GroupHealth Research InstituteSantiago de CompostelaSpain
- Immunology LaboratoryHealth Research InstituteSantiago de CompostelaSpain
| | - José M. Martínez‐Cereijo
- Heart Surgery DepartmentUniversity Hospital of Santiago de CompostelaSantiago de CompostelaSpain
| | - Darío Durán
- Heart Surgery DepartmentUniversity Hospital of Santiago de CompostelaSantiago de CompostelaSpain
- CIBERCV MadridMadridSpain
| | - Moisés Rodríguez‐Mañero
- Translational Cardiology GroupHealth Research InstituteSantiago de CompostelaSpain
- CIBERCV MadridMadridSpain
- Cardiovascular DepartmentUniversity Hospital of Santiago de CompostelaSantiago de CompostelaSpain
| | - José R. González‐Juanatey
- Cardiology GroupHealth Research InstituteSantiago de CompostelaSpain
- CIBERCV MadridMadridSpain
- Cardiovascular DepartmentUniversity Hospital of Santiago de CompostelaSantiago de CompostelaSpain
| | - Sonia Eiras
- Translational Cardiology GroupHealth Research InstituteSantiago de CompostelaSpain
- CIBERCV MadridMadridSpain
| |
Collapse
|
25
|
Ma Q, Immler R, Pruenster M, Sellmayr M, Li C, von Brunn A, von Brunn B, Ehmann R, Wölfel R, Napoli M, Li Q, Romagnani P, Böttcher RT, Sperandio M, Anders HJ, Steiger S. Soluble uric acid inhibits β2 integrin-mediated neutrophil recruitment in innate immunity. Blood 2022; 139:3402-3417. [PMID: 35303071 PMCID: PMC11022987 DOI: 10.1182/blood.2021011234] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/07/2022] [Indexed: 11/20/2022] Open
Abstract
Neutrophils are key players during host defense and sterile inflammation. Neutrophil dysfunction is a characteristic feature of the acquired immunodeficiency during kidney disease. We speculated that the impaired renal clearance of the intrinsic purine metabolite soluble uric acid (sUA) may account for neutrophil dysfunction. Indeed, hyperuricemia (HU, serum UA of 9-12 mg/dL) related or unrelated to kidney dysfunction significantly diminished neutrophil adhesion and extravasation in mice with crystal- and coronavirus-related sterile inflammation using intravital microscopy and an air pouch model. This impaired neutrophil recruitment was partially reversible by depleting UA with rasburicase. We validated these findings in vitro using either neutrophils or serum from patients with kidney dysfunction-related HU with or without UA depletion, which partially normalized the defective migration of neutrophils. Mechanistically, sUA impaired β2 integrin activity and internalization/recycling by regulating intracellular pH and cytoskeletal dynamics, physiological processes that are known to alter the migratory and phagocytic capability of neutrophils. This effect was fully reversible by blocking intracellular uptake of sUA via urate transporters. In contrast, sUA had no effect on neutrophil extracellular trap formation in neutrophils from healthy subjects or patients with kidney dysfunction. Our results identify an unexpected immunoregulatory role of the intrinsic purine metabolite sUA, which contrasts the well-known immunostimulatory effects of crystalline UA. Specifically targeting UA may help to overcome certain forms of immunodeficiency, for example in kidney dysfunction, but may enhance sterile forms of inflammation.
Collapse
Affiliation(s)
- Qiuyue Ma
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Roland Immler
- Walter-Brendel-Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Monika Pruenster
- Walter-Brendel-Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Markus Sellmayr
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Chenyu Li
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Albrecht von Brunn
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University (LMU) Munich and German Center for Infection Research (DZIF), Munich, Germany
| | - Brigitte von Brunn
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University (LMU) Munich and German Center for Infection Research (DZIF), Munich, Germany
| | - Rosina Ehmann
- Bundeswehr Institute of Microbiology, Munich, Germany
| | - Roman Wölfel
- Bundeswehr Institute of Microbiology, Munich, Germany
| | - Matteo Napoli
- Walter-Brendel-Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Qiubo Li
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Paola Romagnani
- Department of Biomedical Experimental and Clinical Sciences “Maria Serio,” University of Florence, Florence, Italy
| | - Ralph Thomas Böttcher
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, Martinsried, Germany
| | - Markus Sperandio
- Walter-Brendel-Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| |
Collapse
|
26
|
Immunosuppressive cells in cancer: mechanisms and potential therapeutic targets. J Hematol Oncol 2022; 15:61. [PMID: 35585567 PMCID: PMC9118588 DOI: 10.1186/s13045-022-01282-8] [Citation(s) in RCA: 265] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/03/2022] [Indexed: 02/08/2023] Open
Abstract
Immunotherapies like the adoptive transfer of gene-engineered T cells and immune checkpoint inhibitors are novel therapeutic modalities for advanced cancers. However, some patients are refractory or resistant to these therapies, and the mechanisms underlying tumor immune resistance have not been fully elucidated. Immunosuppressive cells such as myeloid-derived suppressive cells, tumor-associated macrophages, tumor-associated neutrophils, regulatory T cells (Tregs), and tumor-associated dendritic cells are critical factors correlated with immune resistance. In addition, cytokines and factors secreted by tumor cells or these immunosuppressive cells also mediate the tumor progression and immune escape of cancers. Thus, targeting these immunosuppressive cells and the related signals is the promising therapy to improve the efficacy of immunotherapies and reverse the immune resistance. However, even with certain success in preclinical studies or in some specific types of cancer, large perspectives are unknown for these immunosuppressive cells, and the related therapies have undesirable outcomes for clinical patients. In this review, we comprehensively summarized the phenotype, function, and potential therapeutic targets of these immunosuppressive cells in the tumor microenvironment.
Collapse
|
27
|
Chiscano-Camón L, Plata-Menchaca E, Ruiz-Rodríguez JC, Ferrer R. Fisiopatología del shock séptico. Med Intensiva 2022. [DOI: 10.1016/j.medin.2022.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
28
|
Chiscano-Camón L, Plata-Menchaca E, Ruiz-Rodríguez JC, Ferrer R. [Pathophysiology of septic shock]. Med Intensiva 2022; 46 Suppl 1:1-13. [PMID: 38341256 DOI: 10.1016/j.medine.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/20/2022] [Indexed: 02/12/2024]
Abstract
Sepsis and septic shock result from an inadequate host response to an infection, which causes organ dysfunction. The progression of this condition is manifested by the occurrence of successive clinical stages, resulting from the systemic inflammatory response secondary to the activation of different inflammatory mediators, leading to organ dysfunction. There is a high burden of evidence on the role of endotoxin in the pathogenesis of sepsis and its crucial role in triggering the inflammatory response in sepsis caused by gram-negative bacteria. The coagulation cascade activation in sepsis patients is part of the host's adaptive immune response to infection. The endothelium is the main target in sepsis, which is metabolically active and can.
Collapse
Affiliation(s)
- Luis Chiscano-Camón
- Servicio de Medicina Intensiva, Hospital Universitario Vall d'Hebron, Barcelona, España; Grupo de Investigación Sepsis Organ Dysfunction and Resuscitation (SODIR), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, España; Departament de Medicina. Universitat Autònoma de Barcelona. Barcelona. España
| | - Erika Plata-Menchaca
- Servicio de Medicina Intensiva, Hospital Universitario Vall d'Hebron, Barcelona, España; Grupo de Investigación Sepsis Organ Dysfunction and Resuscitation (SODIR), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, España
| | - Juan Carlos Ruiz-Rodríguez
- Servicio de Medicina Intensiva, Hospital Universitario Vall d'Hebron, Barcelona, España; Grupo de Investigación Sepsis Organ Dysfunction and Resuscitation (SODIR), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, España; Departament de Medicina. Universitat Autònoma de Barcelona. Barcelona. España
| | - Ricard Ferrer
- Servicio de Medicina Intensiva, Hospital Universitario Vall d'Hebron, Barcelona, España; Grupo de Investigación Sepsis Organ Dysfunction and Resuscitation (SODIR), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, España; Departament de Medicina. Universitat Autònoma de Barcelona. Barcelona. España.
| |
Collapse
|
29
|
Stratmann AEP, Wohlgemuth L, Erber ME, Bernhard S, Hug S, Fauler M, Vidoni L, Mohamed AOK, Thomaß BD, Münnich F, Stukan L, Föhr KJ, Mannes M, Huber-Lang MS, Messerer DAC. Simultaneous Measurement of Changes in Neutrophil Granulocyte Membrane Potential, Intracellular pH, and Cell Size by Multiparametric Flow Cytometry. Biomedicines 2021; 9:1504. [PMID: 34829733 PMCID: PMC8614908 DOI: 10.3390/biomedicines9111504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 01/10/2023] Open
Abstract
Neutrophils provide rapid and efficient defense mechanisms against invading pathogens. Upon stimulation with proinflammatory mediators, including complement factors and bacterial peptides, neutrophils respond with changes in their membrane potential, intracellular pH, and cellular size. This study provides an approach to quantify these important changes simultaneously using multiparametric flow cytometry, thereby revealing a typical sequence of neutrophil activation consisting of depolarization, alkalization, and increase in cellular size. Additionally, the time resolution of the flow cytometric measurement is improved in order to allow changes that occur within seconds to be monitored, and thus to enhance the kinetic analysis of the neutrophil response. The method is appropriate for the reliable semiquantitative detection of small variations with respect to an increase, no change, and decrease in those parameters as demonstrated by the screening of various proinflammatory mediators. As a translational outlook, the findings are put into context in inflammatory conditions in vitro as well as in a clinically relevant whole blood model of endotoxemia. Taken together, the multiparametric analysis of neutrophil responsiveness regarding depolarization, alkalization, and changes in cellular size may contribute to a better understanding of neutrophils in health and disease, thus potentially yielding innovative mechanistic insights and possible novel diagnostic and/or prognostic approaches.
Collapse
Affiliation(s)
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Maike Elisabeth Erber
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Stefan Bernhard
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Stefan Hug
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany
| | - Laura Vidoni
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Adam Omar Khalaf Mohamed
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Bertram Dietrich Thomaß
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Frederik Münnich
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Laura Stukan
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, 89081 Ulm, Germany
| | - Karl Josef Föhr
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, 89081 Ulm, Germany
| | - Marco Mannes
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Markus Stefan Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
30
|
Limbu SL, Purba TS, Harries M, Wikramanayake TC, Miteva M, Bhogal RK, O'Neill CA, Paus R. A folliculocentric perspective of dandruff pathogenesis: Could a troublesome condition be caused by changes to a natural secretory mechanism? Bioessays 2021; 43:e2100005. [PMID: 34486144 DOI: 10.1002/bies.202100005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 01/19/2023]
Abstract
Dandruff is a common scalp condition, which frequently causes psychological distress in those affected. Dandruff is considered to be caused by an interplay of several factors. However, the pathogenesis of dandruff remains under-investigated, especially with respect to the contribution of the hair follicle. As the hair follicle exhibits unique immune-modulatory properties, including the creation of an immunoinhibitory, immune-privileged milieu, we propose a novel hypothesis taking into account the role of the hair follicle. We hypothesize that the changes and imbalance of yeast and bacterial species, along with increasing proinflammatory sebum by-products, leads to the activation of immune response and inflammation. Hair follicle keratinocytes may then detect these changes in scalp microbiota resulting in the recruitment of leukocytes to the inflammation site. These changes in the scalp skin immune-microenvironment may impact hair follicle immune privilege status, which opens new avenues into exploring the role of the hair follicle in dandruff pathogenesis. Also see the video abstract here: https://youtu.be/mEZEznCYtNs.
Collapse
Affiliation(s)
- Susan L Limbu
- Centre for Dermatology Research, University of Manchester & NIHR Biomedical Research Centre, Manchester, UK
| | - Talveen S Purba
- Centre for Dermatology Research, University of Manchester & NIHR Biomedical Research Centre, Manchester, UK
| | - Matthew Harries
- Centre for Dermatology Research, University of Manchester & NIHR Biomedical Research Centre, Manchester, UK.,Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Tongyu C Wikramanayake
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mariya Miteva
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ranjit K Bhogal
- Unilever R&D Colworth, Colworth Science Park, Sharnbrook, UK
| | - Catherine A O'Neill
- Centre for Dermatology Research, University of Manchester & NIHR Biomedical Research Centre, Manchester, UK
| | - Ralf Paus
- Centre for Dermatology Research, University of Manchester & NIHR Biomedical Research Centre, Manchester, UK.,Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA.,Monasterium Laboratory, Münster, Germany
| |
Collapse
|
31
|
McCullough LD, Roy-O'Reilly M, Lai YJ, Patrizz A, Xu Y, Lee J, Holmes A, Kraushaar DC, Chauhan A, Sansing LH, Stonestreet BS, Zhu L, Kofler J, Lim YP, Venna VR. Exogenous inter-α inhibitor proteins prevent cell death and improve ischemic stroke outcomes in mice. J Clin Invest 2021; 131:144898. [PMID: 34580244 DOI: 10.1172/jci144898] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Inter-α inhibitor proteins (IAIPs) are a family of endogenous plasma and extracellular matrix molecules. IAIPs suppress proinflammatory cytokines, limit excess complement activation, and bind extracellular histones to form IAIP-histone complexes, leading to neutralization of histone-associated cytotoxicity in models of sepsis. Many of these detrimental processes also play critical roles in the pathophysiology of ischemic stroke. In this study, we first assessed the clinical relevance of IAIPs in stroke and then tested the therapeutic efficacy of exogenous IAIPs in several experimental stroke models. IAIP levels were reduced in both ischemic stroke patients and in mice subjected to experimental ischemic stroke when compared with controls. Post-stroke administration of IAIP significantly improved stroke outcomes across multiple stroke models, even when given 6 hours after stroke onset. Importantly, the beneficial effects of delayed IAIP treatment were observed in both young and aged mice. Using targeted gene expression analysis, we identified a receptor for complement activation, C5aR1, that was highly suppressed in both the blood and brain of IAIP-treated animals. Subsequent experiments using C5aR1-knockout mice demonstrated that the beneficial effects of IAIPs are mediated in part by C5aR1. These results indicate that IAIP is a potential therapeutic candidate for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Louise D McCullough
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Meaghan Roy-O'Reilly
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Yun-Ju Lai
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Anthony Patrizz
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Yan Xu
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Juneyoung Lee
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Aleah Holmes
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Daniel C Kraushaar
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, Texas, USA
| | - Anjali Chauhan
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Lauren H Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Liang Zhu
- Biostatistics and Epidemiology Research Design Core, Center for Clinical and Translational Sciences, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yow-Pin Lim
- ProThera Biologics Inc., Providence, Rhode Island, USA.,Department of Pathology and Laboratory Medicine, The Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Venugopal Reddy Venna
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
32
|
Faget J, Peters S, Quantin X, Meylan E, Bonnefoy N. Neutrophils in the era of immune checkpoint blockade. J Immunother Cancer 2021; 9:jitc-2020-002242. [PMID: 34301813 PMCID: PMC8728357 DOI: 10.1136/jitc-2020-002242] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 01/07/2023] Open
Abstract
The immune checkpoint blockade-based immunotherapies are revolutionizing cancer management. Tumor-associated neutrophils (TANs) were recently highlighted to have a pivotal role in modulating the tumor microenvironment and the antitumor immune response. However, these cells were largely ignored during the development of therapies based on programmed cell death receptor or ligand-1 and cytotoxic T lymphocyte antigen-4 immune checkpoint inhibitors (ICIs). Latest evidences of neutrophil functional diversity in tumor raised many questions and suggest that targeting these cells can offer new treatment opportunities in the context of ICI development. Here, we summarized key information on TAN origin, function, and plasticity that should be considered when developing ICIs and provide a detailed review of the ongoing clinical trials that combine ICIs and a second compound that might affect or be affected by TANs. This review article synthetizes important notions from the literature demonstrating that: (1) Cancer development associates with a profound alteration of neutrophil biogenesis and function that can predict and interfere with the response to ICIs, (2) Neutrophil infiltration in tumor is associated with key features of resistance to ICIs, and (3) TANs play an important role in resistance to antiangiogenic drugs reducing their clinical benefit when used in combination with ICIs. Finally, exploring the clinical/translational aspects of neutrophil impact on the response to ICIs offers the opportunity to propose new translational research avenues to better understand TAN biology and treat patients.
Collapse
Affiliation(s)
- Julien Faget
- IRCM, Inserm, Univ Montpellier, ICM, Montpellier, France, INSERM U1194, Montpellier, France
| | - Solange Peters
- Department of Oncology CHUV-UNIL, University Hospital Lausanne, Lausanne, Switzerland
| | - Xavier Quantin
- Service d'Oncologie Médicale, Institut régional du Cancer de Montpellier, 34298, Montpellier, France
| | - Etienne Meylan
- Swiss Institute for Experimental Cancer Research, EPFL, Lausanne, Switzerland
| | - Nathalie Bonnefoy
- IRCM, Inserm, Univ Montpellier, ICM, Montpellier, France, INSERM U1194, Montpellier, France
| |
Collapse
|
33
|
Reif T, Dyckhoff G, Hohenberger R, Kolbe CC, Gruell H, Klein F, Latz E, Stolp B, Fackler OT. Contact-dependent inhibition of HIV-1 replication in ex vivo human tonsil cultures by polymorphonuclear neutrophils. CELL REPORTS MEDICINE 2021; 2:100317. [PMID: 34195682 PMCID: PMC8233696 DOI: 10.1016/j.xcrm.2021.100317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 03/02/2021] [Accepted: 05/20/2021] [Indexed: 12/01/2022]
Abstract
Polymorphonuclear neutrophils (PMNs), the most abundant white blood cells, are recruited rapidly to sites of infection to exert potent anti-microbial activity. Information regarding their role in infection with human immunodeficiency virus (HIV) is limited. Here we report that addition of PMNs to HIV-infected cultures of human tonsil tissue or peripheral blood mononuclear cells causes immediate and long-lasting suppression of HIV-1 spread and virus-induced depletion of CD4 T cells. This inhibition of HIV-1 spread strictly requires PMN contact with infected cells and is not mediated by soluble factors. 2-Photon (2PM) imaging visualized contacts of PMNs with HIV-1-infected CD4 T cells in tonsil tissue that do not result in lysis or uptake of infected cells. The anti-HIV activity of PMNs also does not involve degranulation, formation of neutrophil extracellular traps, or integrin-dependent cell communication. These results reveal that PMNs efficiently blunt HIV-1 replication in primary target cells and tissue by an unconventional mechanism. PMNs blunt HIV-1 spread and CD4 T cell depletion in HIV-infected human tonsils Suppression of HIV-1 replication by PMNs requires cell-cell contacts PMNs do not affect HIV via effector functions such as NETosis or degranulation PMNs exert unconventional antiviral activity
Collapse
Affiliation(s)
- Tatjana Reif
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Gerhard Dyckhoff
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Ralph Hohenberger
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Carl-Christian Kolbe
- Institute of Innate Immunity, Department of Innate Immunity and Metaflammation, University Hospital Bonn, 53127 Bonn, Germany
| | - Henning Gruell
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Eicke Latz
- Institute of Innate Immunity, Department of Innate Immunity and Metaflammation, University Hospital Bonn, 53127 Bonn, Germany
| | - Bettina Stolp
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Oliver T Fackler
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany.,German Center for Infection Research (DZIF), Partner Site Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
34
|
Jarczak D, Kluge S, Nierhaus A. Sepsis-Pathophysiology and Therapeutic Concepts. Front Med (Lausanne) 2021; 8:628302. [PMID: 34055825 PMCID: PMC8160230 DOI: 10.3389/fmed.2021.628302] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a life-threatening condition and a global disease burden. Today, the heterogeneous syndrome is defined as severe organ dysfunction caused by a dysregulated host response to infection, with renewed emphasis on immune pathophysiology. Despite all efforts of experimental and clinical research during the last three decades, the ability to positively influence course and outcome of the syndrome remains limited. Evidence-based therapy still consists of basic causal and supportive measures, while adjuvant interventions such as blood purification or targeted immunotherapy largely remain without proof of effectiveness so far. With this review, we aim to provide an overview of sepsis immune pathophysiology, to update the choice of therapeutic approaches targeting different immunological mechanisms in the course of sepsis and septic shock, and to call for a paradigm shift from the pathogen to the host response as a potentially more promising angle.
Collapse
Affiliation(s)
- Dominik Jarczak
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel Nierhaus
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
35
|
Silva de França F, Villas-Boas IM, Cogliati B, Woodruff TM, Reis EDS, Lambris JD, Tambourgi DV. C5a-C5aR1 Axis Activation Drives Envenomation Immunopathology by the Snake Naja annulifera. Front Immunol 2021; 12:652242. [PMID: 33936074 PMCID: PMC8082402 DOI: 10.3389/fimmu.2021.652242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/22/2021] [Indexed: 12/03/2022] Open
Abstract
Systemic complement activation drives a plethora of pathological conditions, but its role in snake envenoming remains obscure. Here, we explored complement's contribution to the physiopathogenesis of Naja annulifera envenomation. We found that N. annulifera venom promoted the generation of C3a, C4a, C5a, and the soluble Terminal Complement Complex (sTCC) mediated by the action of snake venom metalloproteinases. N. annulifera venom also induced the release of lipid mediators and chemokines in a human whole-blood model. This release was complement-mediated, since C3/C3b and C5a Receptor 1 (C5aR1) inhibition mitigated the effects. In an experimental BALB/c mouse model of envenomation, N. annulifera venom promoted lipid mediator and chemokine production, neutrophil influx, and swelling at the injection site in a C5a-C5aR1 axis-dependent manner. N. annulifera venom induced systemic complementopathy and increased interleukin and chemokine production, leukocytosis, and acute lung injury (ALI). Inhibition of C5aR1 with the cyclic peptide antagonist PMX205 rescued mice from these systemic reactions and abrogated ALI development. These data reveal hitherto unrecognized roles for complement in envenomation physiopathogenesis, making complement an interesting therapeutic target in envenomation by N. annulifera and possibly by other snake venoms.
Collapse
Affiliation(s)
| | | | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Trent M. Woodruff
- Neuroinflammation Laboratory, School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Edimara da Silva Reis
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | |
Collapse
|
36
|
Bernhard S, Hug S, Stratmann AEP, Erber M, Vidoni L, Knapp CL, Thomaß BD, Fauler M, Nilsson B, Nilsson Ekdahl K, Föhr K, Braun CK, Wohlgemuth L, Huber-Lang M, Messerer DAC. Interleukin 8 Elicits Rapid Physiological Changes in Neutrophils That Are Altered by Inflammatory Conditions. J Innate Immun 2021; 13:225-241. [PMID: 33857948 DOI: 10.1159/000514885] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/22/2021] [Indexed: 11/19/2022] Open
Abstract
A sufficient response of neutrophil granulocytes stimulated by interleukin (IL)-8 is vital during systemic inflammation, for example, in sepsis or severe trauma. Moreover, IL-8 is clinically used as biomarker of inflammatory processes. However, the effects of IL-8 on cellular key regulators of neutrophil properties such as the intracellular pH (pHi) in dependence of ion transport proteins and during inflammation remain to be elucidated. Therefore, we investigated in detail the fundamental changes in pHi, cellular shape, and chemotactic activity elicited by IL-8. Using flow cytometric methods, we determined that the IL-8-induced cellular activity was largely dependent on specific ion channels and transporters, such as the sodium-proton exchanger 1 (NHE1) and non-NHE1-dependent sodium flux. Exposing neutrophils in vitro to a proinflammatory micromilieu with N-formyl-Met-Leu-Phe, LPS, or IL-8 resulted in a diminished response regarding the increase in cellular size and pH. The detailed kinetics of the reduced reactivity of the neutrophil granulocytes could be illustrated in a near-real-time flow cytometric measurement. Last, the LPS-mediated impairment of the IL-8-induced response in neutrophils was confirmed in a translational, animal-free human whole blood model. Overall, we provide novel mechanistic insights for the interaction of IL-8 with neutrophil granulocytes and report in detail about its alteration during systemic inflammation.
Collapse
Affiliation(s)
- Stefan Bernhard
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Stefan Hug
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | | | - Maike Erber
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Laura Vidoni
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Christiane Leonie Knapp
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Bertram Dietrich Thomaß
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, University of Ulm, Ulm, Germany
| | - Bo Nilsson
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristina Nilsson Ekdahl
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Centre of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Karl Föhr
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany
| | - Christian Karl Braun
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany.,Department of Pediatrics and Adolescent Medicine, University Hospital of Ulm, Ulm, Germany
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany.,Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
37
|
Hug S, Bernhard S, Stratmann AEP, Erber M, Wohlgemuth L, Knapp CL, Bauer JM, Vidoni L, Fauler M, Föhr KJ, Radermacher P, Hoffmann A, Huber-Lang M, Messerer DAC. Activation of Neutrophil Granulocytes by Platelet-Activating Factor Is Impaired During Experimental Sepsis. Front Immunol 2021; 12:642867. [PMID: 33796110 PMCID: PMC8007865 DOI: 10.3389/fimmu.2021.642867] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
Platelet-activating factor (PAF) is an important mediator of the systemic inflammatory response. In the case of sepsis, proper activation and function of neutrophils as the first line of cellular defense are based on a well-balanced physiological response. However, little is known about the role of PAF in cellular changes of neutrophils during sepsis. Therefore, this study investigates the reaction patterns of neutrophils induced by PAF with a focus on membrane potential (MP), intracellular pH, and cellular swelling under physiological and pathophysiological conditions and hypothesizes that the PAF-mediated response of granulocytes is altered during sepsis. The cellular response of granulocytes including MP, intracellular pH, cellular swelling, and other activation markers were analyzed by multiparametric flow cytometry. In addition, the chemotactic activity and the formation of platelet-neutrophil complexes after exposure to PAF were investigated. The changes of the (electro-)physiological response features were translationally verified in a human ex vivo whole blood model of endotoxemia as well as during polymicrobial porcine sepsis. In neutrophils from healthy human donors, PAF elicited a rapid depolarization, an intracellular alkalization, and an increase in cell size in a time- and dose-dependent manner. Mechanistically, the alkalization was dependent on sodium-proton exchanger 1 (NHE1) activity, while the change in cellular shape was sodium flux- but only partially NHE1-dependent. In a pathophysiological altered environment, the PAF-induced response of neutrophils was modulated. Acidifying the extracellular pH in vitro enhanced PAF-mediated depolarization, whereas the increases in cell size and intracellular pH were largely unaffected. Ex vivo exposure of human whole blood to lipopolysaccharide diminished the PAF-induced intracellular alkalization and the change in neutrophil size. During experimental porcine sepsis, depolarization of the MP was significantly impaired. Additionally, there was a trend for increased cellular swelling, whereas intracellular alkalization remained stable. Overall, an impaired (electro-)physiological response of neutrophils to PAF stimulation represents a cellular hallmark of those cells challenged during systemic inflammation. Furthermore, this altered response may be indicative of and causative for the development of neutrophil dysfunction during sepsis.
Collapse
Affiliation(s)
- Stefan Hug
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Stefan Bernhard
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | | | - Maike Erber
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Christiane Leonie Knapp
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Jonas Martin Bauer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Laura Vidoni
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, University of Ulm, Ulm, Germany
| | - Karl Josef Föhr
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Development, University Hospital of Ulm, Ulm, Germany
| | - Andrea Hoffmann
- Institute of Anesthesiological Pathophysiology and Process Development, University Hospital of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany.,Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany.,Institute of Anesthesiological Pathophysiology and Process Development, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
38
|
Messerer DAC, Schmidt H, Frick M, Huber-Lang M. Ion and Water Transport in Neutrophil Granulocytes and Its Impairment during Sepsis. Int J Mol Sci 2021; 22:1699. [PMID: 33567720 PMCID: PMC7914618 DOI: 10.3390/ijms22041699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Neutrophil granulocytes are the vanguard of innate immunity in response to numerous pathogens. Their activity drives the clearance of microbe- and damage-associated molecular patterns, thereby contributing substantially to the resolution of inflammation. However, excessive stimulation during sepsis leads to cellular unresponsiveness, immunological dysfunction, bacterial expansion, and subsequent multiple organ dysfunction. During the short lifespan of neutrophils, they can become significantly activated by complement factors, cytokines, and other inflammatory mediators. Following stimulation, the cells respond with a defined (electro-)physiological pattern, including depolarization, calcium influx, and alkalization as well as with increased metabolic activity and polarization of the actin cytoskeleton. Activity of ion transport proteins and aquaporins is critical for multiple cellular functions of innate immune cells, including chemotaxis, generation of reactive oxygen species, and phagocytosis of both pathogens and tissue debris. In this review, we first describe the ion transport proteins and aquaporins involved in the neutrophil ion-water fluxes in response to chemoattractants. We then relate ion and water flux to cellular functions with a focus on danger sensing, chemotaxis, phagocytosis, and oxidative burst and approach the role of altered ion transport protein expression and activity in impaired cellular functions and cell death during systemic inflammation as in sepsis.
Collapse
Affiliation(s)
- David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, 89081 Ulm, Germany;
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, 89081 Ulm, Germany
| | - Hanna Schmidt
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany; (H.S.); (M.F.)
- Department of Pediatrics and Adolescent Medicine, University Hospital of Ulm, 89081 Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany; (H.S.); (M.F.)
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, 89081 Ulm, Germany;
| |
Collapse
|
39
|
Chemical synthesis and characterisation of the complement C5 inhibitory peptide zilucoplan. Amino Acids 2021; 53:143-147. [PMID: 33398524 PMCID: PMC7781173 DOI: 10.1007/s00726-020-02921-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023]
Abstract
The complement component C5 inhibitory peptide zilucoplan is currently in phase III clinical trials for myasthenia gravis (MG). Despite being at an advanced stage of clinical development, there have been no published reports in the literature detailing its chemical synthesis. In this work, we describe an approach for the chemical synthesis of zilucoplan and validate that the synthesised compound blocks LPS-induced C5a production from human blood.
Collapse
|
40
|
Michailidou D, Mustelin T, Lood C. Role of Neutrophils in Systemic Vasculitides. Front Immunol 2020; 11:619705. [PMID: 33391289 PMCID: PMC7774018 DOI: 10.3389/fimmu.2020.619705] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Neutrophils and neutrophil extracellular traps (NETs) contribute to the pathogenesis of many autoimmune diseases, including vasculitis. Though neutrophils, and NETs, can break self-tolerance by being a source of autoantigens for autoantibodies in anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis, playing a key role in driving the autoimmune response, the role of neutrophils and NETs in large vessel vasculitis, including giant cell arteritis (GCA), is not well understood. In this review, we summarize the current insight into molecular mechanisms contributing to neutrophil-mediated pathology in small and medium vessel vasculitis, as well as provide potential translational perspectives on how neutrophils, and NETs, may partake in large vessel vasculitis, a rare disease entity of unclear pathogenesis.
Collapse
Affiliation(s)
- Despina Michailidou
- Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Tomas Mustelin
- Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, WA, United States
| |
Collapse
|
41
|
Messerer DAC, Vidoni L, Erber M, Stratmann AEP, Bauer JM, Braun CK, Hug S, Adler A, Nilsson Ekdahl K, Nilsson B, Barth E, Radermacher P, Huber-Lang M. Animal-Free Human Whole Blood Sepsis Model to Study Changes in Innate Immunity. Front Immunol 2020; 11:571992. [PMID: 33178198 PMCID: PMC7592114 DOI: 10.3389/fimmu.2020.571992] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/21/2020] [Indexed: 12/29/2022] Open
Abstract
Studying innate immunity in humans is crucial for understanding its role in the pathophysiology of systemic inflammation, particularly in the complex setting of sepsis. Therefore, we standardized a step-by-step process from the venipuncture to the transfer in a human model system, while closely monitoring the inflammatory response for up to three hours. We designed an animal-free, human whole blood sepsis model using a commercially available, simple to use, tubing system. First, we analyzed routine clinical parameters, including cell count and blood gas analysis. Second, we demonstrated that extracellular activation markers (e.g., CD11b and CD62l) as well as intracellular metabolic (intracellular pH) and functional (generation of radical oxygen species) features remained stable after incubation in the whole blood model. Third, we mimicked systemic inflammation during early sepsis by exposure of whole blood to pathogen-associated molecular patterns. Stimulation with lipopolysaccharide revealed the capability of the model system to evoke a sepsis-like inflammatory phenotype of innate immunity. In summary, the presented model serves as a convenient, economic, and reliable platform to study innate immunity in human whole blood, which may yield clinically important insights.
Collapse
Affiliation(s)
- David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany.,Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany
| | - Laura Vidoni
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Maike Erber
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | | | - Jonas Martin Bauer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Christian Karl Braun
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Stefan Hug
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Anna Adler
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Kristina Nilsson Ekdahl
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden.,Centre of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Bo Nilsson
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Eberhard Barth
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany
| | - Peter Radermacher
- Institute for Anesthesiologic Pathophysiology and Process Engineering, Ulm University, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
42
|
Nierhaus A, Berlot G, Kindgen-Milles D, Müller E, Girardis M. Best-practice IgM- and IgA-enriched immunoglobulin use in patients with sepsis. Ann Intensive Care 2020; 10:132. [PMID: 33026597 PMCID: PMC7538847 DOI: 10.1186/s13613-020-00740-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 09/06/2020] [Indexed: 12/20/2022] Open
Abstract
Background Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Despite treatment being in line with current guidelines, mortality remains high in those with septic shock. Intravenous immunoglobulins represent a promising therapy to modulate both the pro- and anti-inflammatory processes and can contribute to the elimination of pathogens. In this context, there is evidence of the benefits of immunoglobulin M (IgM)- and immunoglobulin A (IgA)-enriched immunoglobulin therapy for sepsis. This manuscript aims to summarize current relevant data to provide expert opinions on best practice for the use of an IgM- and IgA-enriched immunoglobulin (Pentaglobin) in adult patients with sepsis. Main text Sepsis patients with hyperinflammation and patients with immunosuppression may benefit most from treatment with IgM- and IgA-enriched immunoglobulin (Pentaglobin). Patients with hyperinflammation present with phenotypes that manifest throughout the body, whilst the clinical characteristics of immunosuppression are less clear. Potential biomarkers for hyperinflammation include elevated procalcitonin, interleukin-6, endotoxin activity and C-reactive protein, although thresholds for these are not well-defined. Convenient biomarkers for identifying patients in a stage of immune-paralysis are still matter of debate, though human leukocyte antigen–antigen D related expression on monocytes, lymphocyte count and viral reactivation have been proposed. The timing of treatment is potentially more critical for treatment efficacy in patients with hyperinflammation compared with patients who are in an immunosuppressed stage. Due to the lack of evidence, definitive dosage recommendations for either population cannot be made, though we suggest that patients with hyperinflammation should receive an initial bolus at a rate of up to 0.6 mL (30 mg)/kg/h for 6 h followed by a continuous maintenance rate of 0.2 mL (10 mg)/kg/hour for ≥ 72 h (total dose ≥ 0.9 g/kg). For immunosuppressed patients, dosage is more conservative (0.2 mL [10 mg]/kg/h) for ≥ 72 h, without an initial bolus (total dose ≥ 0.72 g/kg). Conclusions Two distinct populations that may benefit most from Pentaglobin therapy are described in this review. However, further clinical evidence is required to strengthen support for the recommendations given here regarding timing, duration and dosage of treatment.
Collapse
Affiliation(s)
- Axel Nierhaus
- University Medical Center Hamburg, Hamburg, Germany. .,Dep. of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | | | | | | | | |
Collapse
|
43
|
Karasu E, Demmelmaier J, Kellermann S, Holzmann K, Köhl J, Schmidt CQ, Kalbitz M, Gebhard F, Huber-Lang MS, Halbgebauer R. Complement C5a Induces Pro-inflammatory Microvesicle Shedding in Severely Injured Patients. Front Immunol 2020; 11:1789. [PMID: 32983087 PMCID: PMC7492592 DOI: 10.3389/fimmu.2020.01789] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Initially underestimated as platelet dust, extracellular vesicles are continuously gaining interest in the field of inflammation. Various studies addressing inflammatory diseases have shown that microvesicles (MVs) originating from different cell types are systemic transport vehicles carrying distinct cargoes to modulate immune responses. In this study, we focused on the clinical setting of multiple trauma, which is characterized by activation and dysfunction of both, the fluid-phase and the cellular component of innate immunity. Given the sensitivity of neutrophils for the complement anaphylatoxin C5a, we hypothesized that increased C5a production induces alterations in MV shedding of neutrophils resulting in neutrophil dysfunction that fuels posttraumatic inflammation. In a mono-centered prospective clinical study with polytraumatized patients, we found significantly increased granulocyte-derived MVs containing the C5a receptor (C5aR1, CD88) on their surface. This finding was accompanied by a concomitant loss of C5aR1 on granulocytes indicative of an impaired cellular chemotactic and pro-inflammatory neutrophil functions. Furthermore, in vitro exposure of human neutrophils (from healthy volunteers) to C5a significantly increased MV shedding and C5aR1 loss on neutrophils, which could be blocked using the C5aR1 antagonist PMX53. Mechanistic analyses revealed that the interaction between C5aR1 signaling and the small GTPase Arf6 acts as a molecular switch for MV shedding. When neutrophil derived, C5a-induced MV were exposed to a complex ex vivo whole blood model significant pro-inflammatory properties (NADPH activity, ROS and MPO generation) of the MVs became evident. C5a-induced MVs activated resting neutrophils and significantly induced IL-6 secretion. These data suggest a novel role of the C5a-C5aR1 axis: C5a-induced MV shedding from neutrophils results in decreased C5aR1 surface expression on the one hand, on the other hand it leads to profound inflammatory signals which likely are both key drivers of the neutrophil dysfunction which is regularly observed in patients suffering from multiple traumatic injuries.
Collapse
Affiliation(s)
- Ebru Karasu
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Julia Demmelmaier
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Stephanie Kellermann
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Karlheinz Holzmann
- Center for Biomedical Research, Genomics-Core Facility, Ulm University, Ulm, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Lübeck, Germany.,Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - Christoph Q Schmidt
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, Center of Surgery, University of Ulm Medical School, Ulm, Germany
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, Center of Surgery, University of Ulm Medical School, Ulm, Germany
| | - Markus S Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
44
|
Wood AJ, Vassallo AM, Ruchaud-Sparagano MH, Scott J, Zinnato C, Gonzalez-Tejedo C, Kishore K, D'Santos CS, Simpson AJ, Menon DK, Summers C, Chilvers ER, Okkenhaug K, Morris AC. C5a impairs phagosomal maturation in the neutrophil through phosphoproteomic remodeling. JCI Insight 2020; 5:137029. [PMID: 32634128 PMCID: PMC7455072 DOI: 10.1172/jci.insight.137029] [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: 03/02/2020] [Accepted: 06/24/2020] [Indexed: 12/26/2022] Open
Abstract
Critical illness is accompanied by the release of large amounts of the anaphylotoxin, C5a. C5a suppresses antimicrobial functions of neutrophils which is associated with adverse outcomes. The signaling pathways that mediate C5a-induced neutrophil dysfunction are incompletely understood. Healthy donor neutrophils exposed to purified C5a demonstrated a prolonged defect (7 hours) in phagocytosis of Staphylococcus aureus. Phosphoproteomic profiling of 2712 phosphoproteins identified persistent C5a signaling and selective impairment of phagosomal protein phosphorylation on exposure to S. aureus. Notable proteins included early endosomal marker ZFYVE16 and V-ATPase proton channel component ATPV1G1. An assay of phagosomal acidification demonstrated C5a-induced impairment of phagosomal acidification, which was recapitulated in neutrophils from critically ill patients. Examination of the C5a-impaired protein phosphorylation indicated a role for the PI3K VPS34 in phagosomal maturation. Inhibition of VPS34 impaired neutrophil phagosomal acidification and killing of S. aureus. This study provides a phosphoproteomic assessment of human neutrophil signaling in response to S. aureus and its disruption by C5a, identifying a defect in phagosomal maturation and mechanisms of immune failure in critical illness. C5a disrupts the neutrophil phosphoproteomic response to bacteria, impairing phagosomal maturation and bacterial killing.
Collapse
Affiliation(s)
- Alexander Jt Wood
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, United Kingdom
| | - Arlette M Vassallo
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, United Kingdom
| | | | - Jonathan Scott
- Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne, United Kingdom
| | - Carmelo Zinnato
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, United Kingdom
| | - Carmen Gonzalez-Tejedo
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, United Kingdom
| | - Kamal Kishore
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, United Kingdom
| | - Clive S D'Santos
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, United Kingdom
| | - A John Simpson
- Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne, United Kingdom.,Newcastle upon Tyne Hospitals NHS Foundation Trust, Queen Victoria Road, Newcastle upon Tyne, United Kingdom
| | - David K Menon
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, United Kingdom
| | - Charlotte Summers
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, United Kingdom
| | - Edwin R Chilvers
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, United Kingdom.,National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Klaus Okkenhaug
- Division of Immunology, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Andrew Conway Morris
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, United Kingdom.,Division of Immunology, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| |
Collapse
|
45
|
Mollnes TE, Huber-Lang M. Complement in sepsis-when science meets clinics. FEBS Lett 2020; 594:2621-2632. [PMID: 32621378 DOI: 10.1002/1873-3468.13881] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/17/2020] [Accepted: 06/20/2020] [Indexed: 12/18/2022]
Abstract
Sepsis as life-threatening organ dysfunction caused by microorganisms represents a dreadful challenge for the immune system. The role of the complement system as major column of innate immunity has been extensively studied in various sepsis models, but its translational value remains in the dark. Complement activation products, such as C3a and C5a, and their corresponding receptors provide useful diagnostic tools and promising targets to improve organ function and outcome. However, a monotherapeutic complement intervention irrespective of the current immune function seems insufficient to reverse the complex sepsis mechanisms. Indeed, sepsis-induced disturbances of cross talking complement, coagulation, and fibrinolytic cascades lead to systemic 'thromboinflammation', ultimately followed by multiple-organ failure. We propose to reliably monitor the complement function in the patient and to re-establish the immune balance by patient-tailored combined therapies, such as complement and Toll-like receptor inhibition. Our working hypothesis aims at blocking the 'explosive' innate immune recognition systems early on before downstream mediators are released and the inflammatory response becomes irreversible, a strategy that we name 'upstream approach'.
Collapse
Affiliation(s)
- Tom E Mollnes
- Research Laboratory, Nordland Hospital Bodø, Bodø, Norway.,K. G. Jebsen TREC, University of Tromsø, Tromsø, Norway.,Department of Immunology, Oslo University Hospital, and University of Oslo, Oslo, Norway.,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
46
|
Moodley M, Moodley J, Naicker T. The Role of Neutrophils and Their Extracellular Traps in the Synergy of Pre-eclampsia and HIV Infection. Curr Hypertens Rep 2020; 22:41. [PMID: 32462480 DOI: 10.1007/s11906-020-01047-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF THE REVIEW In our innate immune system, neutrophils are the first cells to sense signals of infection and to proceed to kill the invading pathogen. This is mediated by their production of neutrophil extracellular traps (NETS) to entrap pathogenic micro-organisms, preventing their amplification and dissemination. Pre-eclampsia (PE) is the leading cause of global maternal mortality, yet to date, there is no cure nor a gold-standard diagnostic strategy. The purpose of this review is to discover the role of neutrophils in PE as early identification markers. Additionally, this review aims to explore the role of neutrophils in HIV-infected pregnancies with PE as a source of synergy. RECENT FINDINGS Recent findings demonstrate an elevation of neutrophils and neutrophil extracellular traps (NETs) in PE placentae. This is due to their activation by excessive release of syncytiotrophoblast microparticles (STBM). There is also an elevation of NETs in HIV-infected placentae-where histone H3 entraps HIV by binding to its glycoprotein envelope. Additionally, histones H1 and H2A inhibit HIV infection. It is interesting to note that women with both PE and HIV infection have supressed NETs. This review focuses on the role of neutrophils in the synergy of PE and HIV infection. It is plausible that the deregulation of NETs in the synergy of pre-eclamptic HIV-infected women is strategic for the entrapment of the HIV-1 virus. Finally, it is plausible that neutrophils and NETS may act as early biomarkers of PE development. Graphical abstract.
Collapse
Affiliation(s)
- Merantha Moodley
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, College of Health Sciences, Nelson R Mandela School of Medicine, University of Kwa Zulu Natal, Durban, South Africa. .,Discipline of Optics and Imaging, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa.
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, Department of Obstetrics and Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Discipline of Optics and Imaging, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
47
|
Abstract
INTRODUCTION Organ dysfunction remains a major cause of morbidity after trauma. The development of organ dysfunction is determined by the inflammatory response, in which neutrophils are important effector cells. A femoral fracture particularly predisposes for the development of organ dysfunction. This study investigated the chronologic relation between neutrophil characteristics and organ dysfunction in trauma patients with a femoral fracture. METHODS Patients with a femoral fracture presenting at the University Medical Center Utrecht between 2007 and 2013 were included. Data of neutrophil characteristics from standard hematological analyzers were recorded on a daily basis until the 28th day of hospital stay or until discharge. Generalized Estimating Equations were used to compare outcome groups. RESULTS In total 157 patients were analyzed, of whom 81 had polytrauma and 76 monotrauma. Overall mortality within 90 days was 6.4% (n = 10). Eleven patients (7.0%) developed organ dysfunction. In patients who developed organ dysfunction a significant increase in neutrophil count (P = 0.024), a significant increase in neutrophil cell size (P = 0.026), a significant increase in neutrophil complexity (P < 0.004), and a significant decrease in neutrophil lobularity (P < 0.001) were seen after trauma. The rise in neutrophil cell size preceded the clinical manifestation of organ dysfunction in every patient. CONCLUSION Patients who develop organ dysfunction postinjury show changes in neutrophil characteristics before organ dysfunction becomes clinically evident. These findings regarding post-traumatic organ dysfunction may contribute to the development of new prognostic tools for immune-mediated complications in trauma patients. LEVEL OF EVIDENCE Level II, etiologic study.
Collapse
|
48
|
Bashant KR, Toepfner N, Day CJ, Mehta NN, Kaplan MJ, Summers C, Guck J, Chilvers ER. The mechanics of myeloid cells. Biol Cell 2020; 112:103-112. [DOI: 10.1111/boc.201900084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/18/2019] [Accepted: 01/03/2020] [Indexed: 01/05/2023]
Affiliation(s)
- Kathleen R Bashant
- Department of MedicineUniversity of Cambridge Cambridge UK
- Systemic Autoimmunity BranchNational Institute of Arthritis and Musculoskeletal and Skin DiseasesNational Institutes of Health Bethesda Maryland USA
| | - Nicole Toepfner
- Center for Molecular and Cellular BioengineeringBiotechnology Center, Technische Universität Dresden Dresden Germany
- Department of PediatricsUniversity Clinic Carl Gustav Carus, Technische Universität Dresden Dresden Germany
| | | | - Nehal N Mehta
- National Heart Lung and Blood InstituteNational Institutes of Health Bethesda MD USA
| | - Mariana J Kaplan
- Systemic Autoimmunity BranchNational Institute of Arthritis and Musculoskeletal and Skin DiseasesNational Institutes of Health Bethesda Maryland USA
| | | | - Jochen Guck
- Max‐Planck‐Institut für die Physik des Lichts & Max‐Planck‐Zentrum für Physik und Medizin Erlangen Germany
| | | |
Collapse
|
49
|
Patel KJ, Cheng Q, Stephenson S, Allen DP, Li C, Kilkenny J, Finnegan R, Montalvo-Calero V, Esckilsen S, Vasu C, Goddard M, Nadig SN, Atkinson C. Emphysema-associated Autoreactive Antibodies Exacerbate Post-Lung Transplant Ischemia-Reperfusion Injury. Am J Respir Cell Mol Biol 2020; 60:678-686. [PMID: 30571141 DOI: 10.1165/rcmb.2018-0224oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Chronic obstructive pulmonary disease-associated chronic inflammation has been shown to lead to an autoimmune phenotype characterized in part by the presence of lung autoreactive antibodies. We hypothesized that ischemia-reperfusion injury (IRI) liberates epitopes that would facilitate preexisting autoantibody binding, thereby exacerbating lung injury after transplant. We induced emphysema in C57BL/6 mice through 6 months of cigarette smoke (CS) exposure. Mice with CS exposure had significantly elevated serum autoantibodies compared with non-smoke-exposed age-matched (NS) mice. To determine the impact of a full preexisting autoantibody repertoire on IRI, we transplanted BALB/c donor lungs into NS or CS recipients and analyzed grafts 48 hours after transplant. CS recipients had significantly increased lung injury and immune cell infiltration after transplant. Immunofluorescence staining revealed increased IgM, IgG, and C3d deposition in CS recipients. To exclude confounding alloreactivity and confirm the role of preexisting autoantibodies in IRI, syngeneic Rag1-/- (recombination-activating protein 1-knockout) transplants were performed in which recipients were reconstituted with pooled serum from CS or NS mice. Serum from CS-exposed mice significantly increased IRI compared with control mice, with trends in antibody and C3d deposition similar to those seen in allografts. These data demonstrate that pretransplant CS exposure is associated with increased IgM/IgG autoantibodies, which, upon transplant, bind to the donor lung, activate complement, and exacerbate post-transplant IRI.
Collapse
Affiliation(s)
- Kunal J Patel
- 1 Department of Microbiology and Immunology.,2 Lee Patterson Allen Transplant Immunobiology Laboratory, Division of Transplant, Department of Surgery
| | - Qi Cheng
- 1 Department of Microbiology and Immunology.,2 Lee Patterson Allen Transplant Immunobiology Laboratory, Division of Transplant, Department of Surgery.,3 Institute of Organ Transplantation, Department of Surgery, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | | | - D Patterson Allen
- 2 Lee Patterson Allen Transplant Immunobiology Laboratory, Division of Transplant, Department of Surgery
| | - Changhai Li
- 1 Department of Microbiology and Immunology.,2 Lee Patterson Allen Transplant Immunobiology Laboratory, Division of Transplant, Department of Surgery.,3 Institute of Organ Transplantation, Department of Surgery, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Jane Kilkenny
- 2 Lee Patterson Allen Transplant Immunobiology Laboratory, Division of Transplant, Department of Surgery
| | | | | | - Scott Esckilsen
- 2 Lee Patterson Allen Transplant Immunobiology Laboratory, Division of Transplant, Department of Surgery
| | | | - Martin Goddard
- 5 Royal Papworth Hospital NHS Trust, Papworth Everard, Cambridgeshire, United Kingdom
| | - Satish N Nadig
- 1 Department of Microbiology and Immunology.,2 Lee Patterson Allen Transplant Immunobiology Laboratory, Division of Transplant, Department of Surgery.,6 South Carolina Investigators in Transplantation (SCIT), Medical University of South Carolina, Charleston, South Carolina
| | - Carl Atkinson
- 1 Department of Microbiology and Immunology.,2 Lee Patterson Allen Transplant Immunobiology Laboratory, Division of Transplant, Department of Surgery.,6 South Carolina Investigators in Transplantation (SCIT), Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
50
|
Bashant KR, Vassallo A, Herold C, Berner R, Menschner L, Subburayalu J, Kaplan MJ, Summers C, Guck J, Chilvers ER, Toepfner N. Real-time deformability cytometry reveals sequential contraction and expansion during neutrophil priming. J Leukoc Biol 2019; 105:1143-1153. [PMID: 30835869 PMCID: PMC7587463 DOI: 10.1002/jlb.ma0718-295rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 02/08/2019] [Accepted: 02/14/2019] [Indexed: 12/19/2022] Open
Abstract
It has become increasingly apparent that the biomechanical properties of neutrophils impact on their trafficking through the circulation and in particularly through the pulmonary capillary bed. The retention of polarized or shape-changed neutrophils in the lungs was recently proposed to contribute to acute respiratory distress syndrome pathogenesis. Accordingly, this study tested the hypothesis that neutrophil priming is coupled to morpho-rheological (MORE) changes capable of altering cell function. We employ real-time deformability cytometry (RT-DC), a recently developed, rapid, and sensitive way to assess the distribution of size, shape, and deformability of thousands of cells within seconds. During RT-DC analysis, neutrophils can be easily identified within anticoagulated "whole blood" due to their unique granularity and size, thus avoiding the need for further isolation techniques, which affect biomechanical cell properties. Hence, RT-DC is uniquely suited to describe the kinetics of MORE cell changes. We reveal that, following activation or priming, neutrophils undergo a short period of cell shrinking and stiffening, followed by a phase of cell expansion and softening. In some contexts, neutrophils ultimately recover their un-primed mechanical phenotype. The mechanism(s) underlying changes in human neutrophil size are shown to be Na+ /H+ antiport-dependent and are predicted to have profound implications for neutrophil movement through the vascular system in health and disease.
Collapse
Affiliation(s)
- Kathleen R Bashant
- Department of Medicine, University of Cambridge, Cambridge, UK
- National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Reinhard Berner
- Department of Pediatrics, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Leonhard Menschner
- Department of Pediatrics, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | | | | | - Jochen Guck
- Center for Molecular and Cellular Bioengineering, Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | | | - Nicole Toepfner
- Department of Pediatrics, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center for Molecular and Cellular Bioengineering, Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|