1
|
Ji J, Xu X, Zhang L, Liu S, Chen J, Gao H, Xiang L, Li Y, Xu H, Chen Y, Xiang H, Chen S, Han Y, Tang Z, Wang X, Shi X, Mao J, Xi X, Wang J, Fang C. Dedicator of cytokinesis 2 regulates cytoskeletal actin dynamics and is essential for platelet biogenesis and functions. Cardiovasc Res 2025; 121:479-491. [PMID: 39836640 DOI: 10.1093/cvr/cvaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/12/2024] [Indexed: 01/23/2025] Open
Abstract
AIMS Dedicator of cytokinesis 2 (DOCK2), a member of the DOCK family of guanine nucleotide exchange factors that specifically act on the Rho GTPases including Rac and Cdc42, plays pivotal roles in the regulation of leukocyte homeostasis. However, its functions in platelets remain unknown. METHODS AND RESULTS Using mice with genetic deficiency of DOCK2 (Dock2-/-), we showed that Dock2-/- mice exhibited a macrothrombocytopenic phenotype characterized as decreased platelet count and enlarged platelet size by transmission electron microscopy. Dock2-/- megakaryocytes had reduced polyploidization determined by propidium iodide staining and defective proplatelet formation by confocal microscopy. DOCK2 deficiency led to enriched F-actin level in resting platelets but defective F-actin assembly in activated platelets by phalloidin staining, and mechanistically, attenuated activity of Rac1, unchanged Cdc42 but enhanced RhoA measured by immunoprecipitation of GTP-bound proteins. Immunoblotting analysis showed that Dock2-/- platelets had reduced immunoreceptor tyrosine-based activation motif signaling downstream of impaired clustering of GPVI receptors determined by stochastic optical reconstruction microscopy. Further, DOCK2 deficiency resulted in reduced density and branches of fibrin fibres in the clots in vitro and diminished platelet aggregation in a microfluidic chamber ex vivo. Dock2-/- platelets exhibited impaired incorporation into a growing thrombus in cremaster arterioles following allogeneic transfusion into a WT recipient and defective heterotypic interactions with neutrophils in cremaster venules as reflected by decreased platelet-neutrophil aggregate formation in vitro under stirring condition. In addition, myeloid deficiency of DOCK2 caused prolonged tail bleeding times. Finally, pharmacological inhibition of DOCK2 using a small-molecular inhibitor CPYPP suppressed actin dynamics leading to impaired responses to GPVI activation and defects in platelet spreading, clot retraction, and thrombus formation. CONCLUSION DOCK2 plays critical roles in the regulation of platelet biogenesis and functions by controlling Rac1 activity and cytoskeletal actin dynamics and may be a novel target for the treatment of thrombotic and thrombo-inflammatory diseases.
Collapse
Affiliation(s)
- Jiani Ji
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xulin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Wuhan, Hubei 430065, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei 430030, China
- Tongji-Rongcheng Center for Biomedicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lili Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shuang Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiayi Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huihui Gao
- College of Chemistry and Molecular Sciences & Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Limin Xiang
- College of Chemistry and Molecular Sciences & Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Yaofeng Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hui Xu
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yaobing Chen
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Huiqin Xiang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shuai Chen
- Department of Pharmacology, School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, China
| | - Yunyun Han
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhaoming Tang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Department of Pharmacy, Renmin Hospital, Shiyan, Hubei 442000, China
- Biomedical Research Institute, School of Pharmaceutical Sciences and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Xiaofeng Shi
- Department of Hematology, The second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210003, China
| | - Jianhua Mao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaodong Xi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinyu Wang
- Department of Basic Science of Stomatology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, and Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, Hubei 430030, China
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei 430030, China
- Tongji-Rongcheng Center for Biomedicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
2
|
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
|
3
|
Horikawa T, Yagi K, Ishikawa C, Atarashi M, Watanabe A, Kato Y. Continuous renal replacement therapy with vitamin E-coated polysulfone hemofilter reduces inflammatory responses in a porcine lipopolysaccharide-treated model. Ther Apher Dial 2025; 29:96-105. [PMID: 38960621 PMCID: PMC11695089 DOI: 10.1111/1744-9987.14183] [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: 03/27/2024] [Revised: 06/05/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Biological invasions may promote the onset of systemic inflammatory response syndrome in patients eligible for continuous renal replacement therapy (CRRT), leading to poor prognosis. Hence, we aimed to examine the inflammatory reactions in circulation using vitamin E-coated polysulfone hollow fiber membrane (ViLIFE). METHODS Lipopolysaccharides were intravenously administered to pigs (2 μg/kg/30 min) to establish an acute inflammation model. Extracorporeal circulation was performed for 6 h in continuous venovenous hemodiafiltration mode using a hemofilter for CRRT filled with a polysulfone hollow fiber membrane or ViLIFE, and the differences in inflammatory reactions were evaluated. RESULTS The ViLIFE group exhibited low platelet and cytokine levels (p < 0.05 vs. sham-CRRT group). Additionally, the ViLIFE group had lower lactate and high mobility group box 1 levels than the other groups. CONCLUSION ViLIFE represents a promising CRRT modality that can inhibit the inflammatory response in circulation and inhibit further biological invasions.
Collapse
Affiliation(s)
- Takumi Horikawa
- Medical Technology and Material Laboratory, Research and Business Development DivisionAsahi Kasei Medical Co., LTD.ShizuokaJapan
| | - Kana Yagi
- Medical Technology and Material Laboratory, Research and Business Development DivisionAsahi Kasei Medical Co., LTD.ShizuokaJapan
| | - Chika Ishikawa
- Medical Technology and Material Laboratory, Research and Business Development DivisionAsahi Kasei Medical Co., LTD.ShizuokaJapan
| | - Machi Atarashi
- Medical Technology and Material Laboratory, Research and Business Development DivisionAsahi Kasei Medical Co., LTD.ShizuokaJapan
| | - Atsushi Watanabe
- Medical Technology and Material Laboratory, Research and Business Development DivisionAsahi Kasei Medical Co., LTD.ShizuokaJapan
| | - Yoshihisa Kato
- Medical Technology and Material Laboratory, Research and Business Development DivisionAsahi Kasei Medical Co., LTD.ShizuokaJapan
| |
Collapse
|
4
|
Kumar V, Stewart Iv JH. Platelet's plea to Immunologists: Please do not forget me. Int Immunopharmacol 2024; 143:113599. [PMID: 39547015 DOI: 10.1016/j.intimp.2024.113599] [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: 07/09/2024] [Revised: 10/07/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Platelets are non-nucleated mammalian cells originating from the cytoplasmic expulsion of the megakaryocytes. Megakaryocytes develop during hematopoiesis through megakaryopoiesis, whereas platelets develop from megakaryocytes through thrombopoiesis. Since their first discovery, platelets have been studied as critical cells controlling hemostasis or blood coagulation. However, coagulation and innate immune response are evolutionarily linked processes. Therefore, it has become critical to investigate the immunological functions of platelets to maintain immune homeostasis. Advances in immunology and platelet biology research have explored different critical roles of platelets, including phagocytosis, release of different immune mediators, and controlling functions of different immune cells by direct interaction and immune mediators. The current article discusses platelet's development and their critical role as innate immune cells, which express different pattern recognition receptors (PRRs), recognizing different pathogen or microbe-associated molecular patterns (PAMPs or MAMPs) and death/damage-associated molecular patterns (DAMPs) and their direct interactions with innate and adaptive immune cells to maintain immune homeostasis.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Medical Education Building-C, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310 USA.
| | - John H Stewart Iv
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Medical Education Building-C, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310 USA
| |
Collapse
|
5
|
Ye W, Xu S, Liu Y, Ye Z. Role of endothelial glycocalyx in central nervous system diseases and evaluation of the targeted therapeutic strategies for its protection: a review of clinical and experimental data. Rev Neurosci 2024; 35:839-853. [PMID: 39034663 DOI: 10.1515/revneuro-2024-0039] [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: 03/14/2024] [Accepted: 05/22/2024] [Indexed: 07/23/2024]
Abstract
Central nervous system (CNS) diseases, such as stroke, traumatic brain injury, dementia, and demyelinating diseases, are generally characterized by high morbidity and mortality, which impose a heavy economic burden on patients and their caregivers throughout their lives as well as on public health. The occurrence and development of CNS diseases are closely associated with a series of pathophysiological changes including inflammation, blood-brain barrier disruption, and abnormal coagulation. Endothelial glycocalyx (EG) plays a key role in these changes, making it a novel intervention target for CNS diseases. Herein, we review the current understanding of the role of EG in common CNS diseases, from the perspective of individual pathways/cytokines in pathophysiological and systematic processes. Furthermore, we emphasize the recent developments in therapeutic agents targeted toward protection or restoration of EG. Some of these treatments have yielded unexpected pharmacological results, as previously unknown mechanisms underlying the degradation and destruction of EG has been brought to light. Furthermore, the anti-inflammatory, anticoagulative, and antioxidation effects of EG and its protective role exerted via the blood-brain barrier have been recognized.
Collapse
Affiliation(s)
- Weihao Ye
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Shang Xu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ying Liu
- Department of Rehabilitation Medicine, 117742The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ziming Ye
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| |
Collapse
|
6
|
Peshkova AD, Saliakhutdinova SM, Sounbuli K, Selivanova YA, Andrianova IA, Khabirova AI, Litvinov RI, Weisel JW. The differential formation and composition of leukocyte-platelet aggregates induced by various cellular stimulants. Thromb Res 2024; 241:109092. [PMID: 39024901 PMCID: PMC11411814 DOI: 10.1016/j.thromres.2024.109092] [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: 05/01/2024] [Revised: 06/16/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Leukocyte-platelet aggregates comprise a pathogenic link between hemostasis and immunity, but the prerequisites and mechanisms of their formation remain not understood. AIMS To quantify the formation, composition, and morphology of leukocyte-platelet aggregates in vitro under the influence of various cellular activators. METHODS Phorbol-12-myristate-13-acetate (PMA), lipopolysaccharide (LPS), thrombin receptor-activating peptide (TRAP-6), and adenosine diphosphate (ADP) were used as cellular activators. Flow cytometry was utilized to identify and quantify aggregates in whole human blood and platelet-rich plasma. Cell types and cellular aggregates were identified using fluorescently labeled antibodies against the appropriate cellular markers, and cell activation was assessed by the expression of appropriate surface markers. For confocal fluorescent microscopy, cell membranes and nuclei were labeled. Neutrophil-platelet aggregates were studied using scanning electron microscopy. RESULTS In the presence of PMA, ADP or TRAP-6, about 17-38 % of neutrophils and 61-77 % of monocytes formed aggregates with platelets in whole blood, whereas LPS did not induce platelet aggregation with either neutrophils or monocytes due the inability to activate platelets. Similar results were obtained when isolated neutrophils were added to platelet-rich plasma. All the cell types involved in the heterotypic aggregation expressed molecular markers of activation. Fluorescent and electron microscopy of the aggregates showed that the predominant platelet/leukocyte ratios were 1:1 and 2:1. CONCLUSIONS Formation of leukocyte-platelet aggregates depends on the nature of the cellular activator and the spectrum of its cell-activating ability. An indispensable condition for formation of leukocyte-platelet aggregates is activation of all cell types including platelets, which is the restrictive step.
Collapse
Affiliation(s)
- Alina D Peshkova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | | | - Khetam Sounbuli
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Yuliya A Selivanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Izabella A Andrianova
- Department of Internal Medicine, Division of Hematology and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Alina I Khabirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Rustem I Litvinov
- Departments of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - John W Weisel
- Departments of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Chen WA, Boskovic DS. Neutrophil Extracellular DNA Traps in Response to Infection or Inflammation, and the Roles of Platelet Interactions. Int J Mol Sci 2024; 25:3025. [PMID: 38474270 DOI: 10.3390/ijms25053025] [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: 11/30/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Neutrophils present the host's first line of defense against bacterial infections. These immune effector cells are mobilized rapidly to destroy invading pathogens by (a) reactive oxygen species (ROS)-mediated oxidative bursts and (b) via phagocytosis. In addition, their antimicrobial service is capped via a distinct cell death mechanism, by the release of their own decondensed nuclear DNA, supplemented with a variety of embedded proteins and enzymes. The extracellular DNA meshwork ensnares the pathogenic bacteria and neutralizes them. Such neutrophil extracellular DNA traps (NETs) have the potential to trigger a hemostatic response to pathogenic infections. The web-like chromatin serves as a prothrombotic scaffold for platelet adhesion and activation. What is less obvious is that platelets can also be involved during the initial release of NETs, forming heterotypic interactions with neutrophils and facilitating their responses to pathogens. Together, the platelet and neutrophil responses can effectively localize an infection until it is cleared. However, not all microbial infections are easily cleared. Certain pathogenic organisms may trigger dysregulated platelet-neutrophil interactions, with a potential to subsequently propagate thromboinflammatory processes. These may also include the release of some NETs. Therefore, in order to make rational intervention easier, further elucidation of platelet, neutrophil, and pathogen interactions is still needed.
Collapse
Affiliation(s)
- William A Chen
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University, Loma Linda, CA 92350, USA
| | - Danilo S Boskovic
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
- Department of Earth and Biological Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| |
Collapse
|
8
|
Carestia A, Godin LC, Jenne CN. Step up to the platelet: Role of platelets in inflammation and infection. Thromb Res 2023; 231:182-194. [PMID: 36307228 DOI: 10.1016/j.thromres.2022.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022]
Abstract
Platelets are anucleated cells derived from megakaryocytes that are primarily responsible for hemostasis. However, in recent years, these cytoplasts have become increasingly recognized as immune cells, able to detect, interact with, and kill pathogens. As platelets are involved in both immunity and coagulation, they have a central role in immunothrombosis, a physiological process in which immune cells induce the formation of microthrombi to both prevent the spread of pathogens, and to help facilitate clearance. In this review, we will highlight the role of platelets as key players in the inflammatory and innate immune response against bacterial and viral infection, including direct and indirect interactions with pathogens and other immune cells.
Collapse
Affiliation(s)
- Agostina Carestia
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| | - Laura C Godin
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| | - Craig N Jenne
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| |
Collapse
|
9
|
Schönichen C, Montague SJ, Brouns SL, Burston JJ, Cosemans JM, Jurk K, Kehrel BE, Koenen RR, Ní Áinle F, O’Donnell VB, Soehnlein O, Watson SP, Kuijpers MJ, Heemskerk JW, Nagy M. Antagonistic Roles of Human Platelet Integrin αIIbβ3 and Chemokines in Regulating Neutrophil Activation and Fate on Arterial Thrombi Under Flow. Arterioscler Thromb Vasc Biol 2023; 43:1700-1712. [PMID: 37409530 PMCID: PMC10443630 DOI: 10.1161/atvbaha.122.318767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Platelets and neutrophils are the first blood cells accumulating at sites of arterial thrombus formation, and both cell types contribute to the pathology of thrombotic events. We aimed to identify key interaction mechanisms between these cells using microfluidic approaches. METHODS Whole-blood perfusion was performed over a collagen surface at arterial shear rate. Platelet and leukocyte (in majority neutrophil) activation were microscopically visualized using fluorescent markers. The contributions of platelet-adhesive receptors (integrin, P-selectin, CD40L) and chemokines were studied by using inhibitors or antibodies and using blood from patients with GT (Glanzmann thrombasthenia) lacking platelet-expressed αIIbβ3. RESULTS We observed (1) an unknown role of activated platelet integrin αIIbß3 preventing leukocyte adhesion, which was overcome by short-term flow disturbance provoking massive adhesion; (2) that platelet-expressed CD40L controls the crawling pattern and thrombus fidelity of the cells on a thrombus; (3) that continued secretion of platelet substances promotes activation of identified neutrophils, as assessed by (fMLP [N-formylmethionyl-leucyl-phenylalanine, a potent chemotactic agent and leukocyte activator] induced) [Ca2+]i rises and antigen expression; (4) and that platelet-released chemokines activate the adhered cells in the order of CXCL7>CCL5>CXCL4. Furthermore, postsilencing of the platelets in a thrombus suppressed the leukocyte activation. However, the leukocytes on thrombi did no more than limitedly form neutrophil extracellular traps, unless stimulated with phorbol ester or lipopolysaccharide. CONCLUSIONS Together, these findings reveal a multifaceted regulation of adhesion and activation of neutrophils by platelets in a thrombus, with a balanced role of several platelet-adhesive receptors and a promoting role of platelet-released substances. This multivalent nature of neutrophil-thrombus interactions offers novel prospects for pharmacological intervention.
Collapse
Affiliation(s)
- Claudia Schönichen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany (C.S., K.J.)
| | - Samantha J. Montague
- Institute of Cardiovascular Sciences, The Medical School, University of Birmingham, United Kingdom (S.J.M., S.P.W.)
| | - Sanne L.N. Brouns
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
| | - James J. Burston
- Systems Immunity Research Institute, School of Medicine, Cardiff University, United Kingdom (J.J.B., V.B.O.)
| | - Judith M.E.M. Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany (C.S., K.J.)
- Department of Anaesthesiology and Intensive Care, University Hospital Muenster, Germany (K.J., B.E.K.)
| | - Beate E. Kehrel
- Department of Anaesthesiology and Intensive Care, University Hospital Muenster, Germany (K.J., B.E.K.)
| | - Rory R. Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
| | - Fionnuala Ní Áinle
- School of Medicine, University College Dublin, Ireland (F.N.Á.)
- Department of Haematology, Mater Misericordiae University Hospital and Rotunda Hospital, Dublin, Ireland (F.N.Á.)
| | - Valerie B. O’Donnell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, United Kingdom (J.J.B., V.B.O.)
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Germany (O.S.)
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Westfälische Wilhelms Universität, Münster, Germany (O.S.)
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (O.S.)
| | - Steve P. Watson
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
- Institute of Cardiovascular Sciences, The Medical School, University of Birmingham, United Kingdom (S.J.M., S.P.W.)
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, the Midlands, United Kingdom (S.P.W.)
| | - Marijke J.E. Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
- Thrombosis Expertise Centre, Heart and Vascular Centre, Maastricht University Medical Centre, the Netherlands (M.J.E.K.)
| | - Johan W.M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
- Synapse Research Institute, Maastricht, the Netherlands (J.W.M.H.)
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
| |
Collapse
|
10
|
Tang C, Chen G, Wu F, Cao Y, Yang F, You T, Liu C, Li M, Hu S, Ren L, Lu Q, Deng W, Xu Y, Wang G, Jo H, Zhang Y, Wu Y, Zabel BA, Zhu L. Endothelial CCRL2 induced by disturbed flow promotes atherosclerosis via chemerin-dependent β2 integrin activation in monocytes. Cardiovasc Res 2023; 119:1811-1824. [PMID: 37279540 PMCID: PMC10405567 DOI: 10.1093/cvr/cvad085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/01/2023] [Indexed: 06/08/2023] Open
Abstract
AIMS Chemoattractants and their cognate receptors are essential for leucocyte recruitment during atherogenesis, and atherosclerotic plaques preferentially occur at predilection sites of the arterial wall with disturbed flow (d-flow). In profiling the endothelial expression of atypical chemoattractant receptors (ACKRs), we found that Ackr5 (CCRL2) was up-regulated in an endothelial subpopulation by atherosclerotic stimulation. We therefore investigated the role of CCRL2 and its ligand chemerin in atherosclerosis and the underlying mechanism. METHODS AND RESULTS By analysing scRNA-seq data of the left carotid artery under d-flow and scRNA-seq datasets GSE131776 of ApoE-/- mice from the Gene Expression Omnibus database, we found that CCRL2 was up-regulated in one subpopulation of endothelial cells in response to d-flow stimulation and atherosclerosis. Using CCRL2-/-ApoE-/- mice, we showed that CCRL2 deficiency protected against plaque formation primarily in the d-flow areas of the aortic arch in ApoE-/- mice fed high-fat diet. Disturbed flow induced the expression of vascular endothelial CCRL2, recruiting chemerin, which caused leucocyte adhesion to the endothelium. Surprisingly, instead of binding to monocytic CMKLR1, chemerin was found to activate β2 integrin, enhancing ERK1/2 phosphorylation and monocyte adhesion. Moreover, chemerin was found to have protein disulfide isomerase-like enzymatic activity, which was responsible for the interaction of chemerin with β2 integrin, as identified by a Di-E-GSSG assay and a proximity ligation assay. For clinical relevance, relatively high serum levels of chemerin were found in patients with acute atherothrombotic stroke compared to healthy individuals. CONCLUSIONS Our findings indicate that d-flow-induced CCRL2 promotes atherosclerotic plaque formation via a novel CCRL2-chemerin-β2 integrin axis, providing potential targets for the prevention or therapeutic intervention of atherosclerosis.
Collapse
Affiliation(s)
- Chaojun Tang
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, China
- JinFeng Laboratory, Chongqing, China
| | - Guona Chen
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Fan Wu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Cambridge-Suda Genomic Resource Center, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Yiren Cao
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Fei Yang
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Tao You
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chu Liu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Menglu Li
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Shuhong Hu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Lijie Ren
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Qiongyu Lu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Wei Deng
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Ying Xu
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Cambridge-Suda Genomic Resource Center, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Guixue Wang
- JinFeng Laboratory, Chongqing, China
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Yonghong Zhang
- Department of Epidemiology School of Public Health, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
| | - Yi Wu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Brian A Zabel
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Li Zhu
- Cyrus Tang Medical Institute, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- The Ninth Affiliated Hospital, Soochow University, Rm 509, Bldg 703, 199 Ren’ai Road, Suzhou 215123, China
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, China
- JinFeng Laboratory, Chongqing, China
| |
Collapse
|
11
|
Dhanesha N, Ansari J, Pandey N, Kaur H, Virk C, Stokes KY. Poststroke venous thromboembolism and neutrophil activation: an illustrated review. Res Pract Thromb Haemost 2023; 7:100170. [PMID: 37274177 PMCID: PMC10236222 DOI: 10.1016/j.rpth.2023.100170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 06/06/2023] Open
Abstract
Patients with acute ischemic stroke are at a high risk of venous thromboembolism (VTE), such as deep vein thrombosis (DVT), estimated to affect approximately 80,000 patients with stroke each year in the United States. The prevalence of symptomatic DVT after acute stroke is approximately 10%. VTE is associated with increased rates of in-hospital death and disability, with higher prevalence of in-hospital complications and increased 1-year mortality in patients with stroke. Current guidelines recommend the use of pharmacologic VTE prophylaxis in patients with acute ischemic stroke. However, thromboprophylaxis prevents only half of expected VTE events and is associated with high risk of bleeding, suggesting the need for targeted alternative treatments to reduce VTE risk in these patients. Neutrophils are among the first cells in blood to respond after ischemic stroke. Importantly, coordinated interactions among neutrophils, platelets, and endothelial cells contribute to the development of DVT. In case of stroke and other related immune disorders, such as antiphospholipid syndrome, neutrophils potentiate thrombus propagation through the formation of neutrophil-platelet aggregates, secreting inflammatory mediators, complement activation, releasing tissue factor, and producing neutrophil extracellular traps. In this illustrated review article, we present epidemiology and management of poststroke VTE, preclinical and clinical evidence of neutrophil hyperactivation in stroke, and mechanisms for neutrophil-mediated VTE in the context of stroke. Given the hyperactivation of circulating neutrophils in patients with stroke, we propose that a better understanding of molecular mechanisms leading to neutrophil activation may result in the development of novel therapeutics to reduce the risk of VTE in this patient population.
Collapse
Affiliation(s)
- Nirav Dhanesha
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Junaid Ansari
- Department of Neurology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Nilesh Pandey
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Harpreet Kaur
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Chiranjiv Virk
- Division of Vascular Surgery and Endovascular Surgery, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Karen Y. Stokes
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| |
Collapse
|
12
|
Banka AL, Guevara MV, Brannon ER, Nguyen NQ, Song S, Cady G, Pinsky DJ, Uhrich KE, Adili R, Holinstat M, Eniola-Adefeso O. Cargo-free particles divert neutrophil-platelet aggregates to reduce thromboinflammation. Nat Commun 2023; 14:2462. [PMID: 37117163 PMCID: PMC10144907 DOI: 10.1038/s41467-023-37990-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 04/11/2023] [Indexed: 04/30/2023] Open
Abstract
The combination of inflammation and thrombosis is a hallmark of many cardiovascular diseases. Under such conditions, platelets are recruited to an area of inflammation by forming platelet-leukocyte aggregates via interaction of PSGL-1 on leukocytes and P-selectin on activated platelets, which can bind to the endothelium. While particulate drug carriers have been utilized to passively redirect leukocytes from areas of inflammation, the downstream impact of these carriers on platelet accumulation in thromboinflammatory conditions has yet to be studied. Here, we explore the ability of polymeric particles to divert platelets away from inflamed blood vessels both in vitro and in vivo. We find that untargeted and targeted micron-sized polymeric particles can successfully reduce platelet adhesion to an inflamed endothelial monolayer in vitro in blood flow systems and in vivo in a lipopolysaccharide-induced, systemic inflammation murine model. Our data represent initial work in developing cargo-free, anti-platelet therapeutics specifically for conditions of thromboinflammation.
Collapse
Affiliation(s)
- Alison L Banka
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - M Valentina Guevara
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Emma R Brannon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nhien Q Nguyen
- Department of Chemistry, University of California Riverside, Riverside, CA, 92521, USA
| | - Shuang Song
- Department of Chemistry, University of California Riverside, Riverside, CA, 92521, USA
| | - Gillian Cady
- Division of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David J Pinsky
- Division of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kathryn E Uhrich
- Department of Chemistry, University of California Riverside, Riverside, CA, 92521, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Holinstat
- Division of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
13
|
Bruserud Ø, Mosevoll KA, Bruserud Ø, Reikvam H, Wendelbo Ø. The Regulation of Neutrophil Migration in Patients with Sepsis: The Complexity of the Molecular Mechanisms and Their Modulation in Sepsis and the Heterogeneity of Sepsis Patients. Cells 2023; 12:cells12071003. [PMID: 37048076 PMCID: PMC10093057 DOI: 10.3390/cells12071003] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Common causes include gram-negative and gram-positive bacteria as well as fungi. Neutrophils are among the first cells to arrive at an infection site where they function as important effector cells of the innate immune system and as regulators of the host immune response. The regulation of neutrophil migration is therefore important both for the infection-directed host response and for the development of organ dysfunctions in sepsis. Downregulation of CXCR4/CXCL12 stimulates neutrophil migration from the bone marrow. This is followed by transmigration/extravasation across the endothelial cell barrier at the infection site; this process is directed by adhesion molecules and various chemotactic gradients created by chemotactic cytokines, lipid mediators, bacterial peptides, and peptides from damaged cells. These mechanisms of neutrophil migration are modulated by sepsis, leading to reduced neutrophil migration and even reversed migration that contributes to distant organ failure. The sepsis-induced modulation seems to differ between neutrophil subsets. Furthermore, sepsis patients should be regarded as heterogeneous because neutrophil migration will possibly be further modulated by the infecting microorganisms, antimicrobial treatment, patient age/frailty/sex, other diseases (e.g., hematological malignancies and stem cell transplantation), and the metabolic status. The present review describes molecular mechanisms involved in the regulation of neutrophil migration; how these mechanisms are altered during sepsis; and how bacteria/fungi, antimicrobial treatment, and aging/frailty/comorbidity influence the regulation of neutrophil migration.
Collapse
Affiliation(s)
- Øystein Bruserud
- Leukemia Research Group, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence:
| | - Knut Anders Mosevoll
- Section for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Section for Infectious Diseases, Department of Clinical Research, University of Bergen, 5021 Bergen, Norway
| | - Øyvind Bruserud
- Department for Anesthesiology and Intensive Care, Haukeland University Hospital, 5021 Bergen, Norway
| | - Håkon Reikvam
- Leukemia Research Group, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Øystein Wendelbo
- Section for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Faculty of Health, VID Specialized University, Ulriksdal 10, 5009 Bergen, Norway
| |
Collapse
|
14
|
Platelet activation and ferroptosis mediated NETosis drives heme induced pulmonary thrombosis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166688. [PMID: 36925054 DOI: 10.1016/j.bbadis.2023.166688] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Cell-free heme (CFH) is a product of hemoglobin, myoglobin and hemoprotein degradation, which is a hallmark of pathologies associated with extensive hemolysis and tissue damage. CHF and iron collectively induce cytokine storm, lung injury, respiratory distress and infection susceptibility in the lungs suggesting their key role in the progression of lung disease pathology. We have previously demonstrated that heme-mediated reactive oxygen species (ROS) induces platelet activation and ferroptosis. However, interaction of ferroptotic platelets and neutrophils, the mechanism of action and associated complications remain unclear. In this study, we demonstrate that heme-induced P-selectin expression and Phosphatidylserine (PS) externalization in platelets via ASK-1-inflammasome axis increases platelet-neutrophil aggregates in circulation, resulting in Neutrophil extracellular traps (NET) formation in vitro and in vivo. Further, heme-induced platelet activation in mice increased platelet-neutrophil aggregates and accumulation of NETs in the lungs causing pulmonary damage. Thus, connecting CFH-mediated platelet activation to NETosis and pulmonary thrombosis. As lung infections induce acute respiratory stress, thrombosis and NETosis, we propose that heme -mediated platelet activation and ferroptosis might be crucial in such clinical manifestations. Further, considering the ability of redox modulators and ferroptosis inhibitors like FS-1, Lpx-1 and DFO to inhibit heme-induced ferroptotic platelets-mediated NETosis and pulmonary thrombosis. They could be potential adjuvant therapy to regulate respiratory distress-associated clinical complications.
Collapse
|
15
|
Segalo S, Kiseljakovic E, Papic E, Joguncic A, Pasic A, Sahinagic M, Lepara O, Sporisevic L. The Role of Hemogram-derived Ratios in COVID-19 Severity Stratification in a Primary Healthcare Facility. Acta Inform Med 2023; 31:41-47. [PMID: 37038490 PMCID: PMC10082658 DOI: 10.5455/aim.2023.31.41-47] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/15/2023] [Indexed: 04/12/2023] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) can cause a wide clinical spectrum, ranging from asymptomatic to severe disease with a high mortality rate. In view of the current pandemic and the increasing influx of patients into healthcare facilities, there is a need to identify simple and reliable tools for stratifying patients. Objective Study aimed to analyze whether hemogram-derived ratios (HDRs) can be used to identify patients with a risk of developing a severe clinical form and admission to hospital. Methods This cross-sectional and observational study included 500 patients with a confirmed diagnosis of COVID-19. Data on clinical features and laboratory parameters were collected from medical records and 13 HDRs were calculated and analyzed. Descriptive and inferential statistics were included in the analysis. Results Of the 500 patients, 43.8% had a severe form of the disease. Lymphocytopenia, monocytopenia, higher C-reactive protein (CRP), and erythrocyte sedimentation rate (ESR) were found in severe patients (p < 0.05). Significantly higher neutrophil-to-lymphocyte ratio (NLR), derived NLR (dNLR), neutrophil-to-platelet ratio (NPR), neutrophil-to-lymphocyte-to-platelet ratio (NLPR) and CRP-to-lymphocyte ratio (CRP/Ly) values were found in severe patients (p < 0.001). In addition, they have statistically significant prognostic potential (p < 0.001). The area under the curve (AUC) for CRP/Ly, dNLR, NLPR, NLR, and NPR were 0.693, 0.619, 0.619, 0.616, and 0.603, respectively. The sensitivity and specificity were 65.7% and 65.6% for CRP/Ly, 51.6% and 70.8 for dNLR, 61.6% and 57.3% for NLPR, 40.6% and 80.4% for NLR, and 48.8% and 69.1% for NPR. Conclusion The results of the study suggest that NLR, dNLR, CRP/Ly, NPR, and NLPR can be considered as potentially useful markers for stratifying patients with a severe form of the disease. HDRs derived from routine blood tests results should be included in common laboratory practice since they are readily available, easy to calculate, and inexpensive.
Collapse
Affiliation(s)
- Sabina Segalo
- University of Sarajevo, Faculty of Health Studies, Sarajevo, Bosnia and Herzegovina
| | - Emina Kiseljakovic
- University of Sarajevo, Faculty of Medicine, Sarajevo, Bosnia and Herzegovina
| | - Emsel Papic
- University of Sarajevo, Faculty of Health Studies, Sarajevo, Bosnia and Herzegovina
| | - Anes Joguncic
- University of Sarajevo, Faculty of Health Studies, Sarajevo, Bosnia and Herzegovina
- Public Health Institute of Canton Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Aleksandra Pasic
- University of Sarajevo, Faculty of Health Studies, Sarajevo, Bosnia and Herzegovina
- Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Mubera Sahinagic
- Public Institution Medical Center of Sarajevo Canton, Sarajevo, Bosnia and Herzegovina
| | - Orhan Lepara
- University of Sarajevo, Faculty of Medicine, Sarajevo, Bosnia and Herzegovina
| | - Lutvo Sporisevic
- Public Institution Medical Center of Sarajevo Canton, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
16
|
Selim ZI, Mostafa NM, Ismael EO, Kamal D. Platelet lymphocyte ratio, lymphocyte monocyte ratio, mean platelet volume, and neutrophil lymphocyte ratio in Behcet’s disease and their relation to disease activity. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2023. [DOI: 10.1186/s43166-022-00168-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Abstract
Background
Behcet’s disease (BD) does not have specific laboratory finding or pathological physical examination sign, and only few studies have investigated Neutrophil to lymphocyte ratio (NLR), platelets to lymphocytes ratio (PLR), lymphocytes to monocytes ratio (LMR), or mean platelet volume (MPV) values in patients with BD. We conducted this study to investigate the relationship between these indices and Behcet’s disease (BD) and to determine their relation to BD disease activity.
Results
This study is a case-control study that included 36 Behcet’s disease patients and 36 healthy controls. BD patients showed significant increase in the mean of NLR and PLR in comparison to control (P = 0.008 and 0.011) respectively, and highly significant decrease in LMR and MPV levels in BD patients in comparison to control (P < 0.001 and < 0.001) respectively. Also, we found that NLR, PLR, and LMR were significantly related to BD activity, and there were significant associations between the studied hematological parameters with some of muco-cutaneous, articular, gastrointestinal, eye, and nervous system manifestations in BD patients.
Conclusion
The blood indices NLR, PLR, LMR, and MPV are potential inflammatory markers that can be used to evaluate inflammatory status and disease activity in patients with BD. NLR and PLR showed positive relation being higher in active disease and also higher in highly active disease than in low disease activity. Also, LMR was significantly decreased in Behcet’s disease patients in relation to disease activity. Furthermore, NLR and PLR levels were significantly more associated with muco-cutaneous and nervous system involvement while, LMR levels were significantly associated with muco-cutaneous, articular, gastrointestinal and eye manifestations and MPV levels were associated with articular manifestations being significantly related to disease activity. These easily evaluated markers could help in the management of this disease with multisystem affection that are sometimes serious and potentially life threatening.
Collapse
|
17
|
Kim HK, Lee KO, Oh SH, Lee KY, Choo SW, Kim OJ, Kim TG, Kim SH, Na SJ, Heo JH. The clinical significance of peripheral blood cell ratios in patients with intracranial aneurysm. Front Neurol 2022; 13:1080244. [PMID: 36605785 PMCID: PMC9807666 DOI: 10.3389/fneur.2022.1080244] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Background and objective Inflammation is an important factor in the development of aneurysm, and has been identified as a key characteristic predictive of rupture of intracranial aneurysm (IA). However, the role of inflammatory peripheral blood cell ratios in patients with IA has not been well delineated. Methods A total of 1,209 patients, including 1,001 with unruptured IA and 208 with ruptured IA, were enrolled in this study. Neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), platelet-to-neutrophil ratio (PNR), and platelet-to-white-blood-cell ratio (PWR) were compared between ruptured and unruptured IA. Results Compared with the ruptured IA group, the unruptured IA group had higher PNR {median, 65.96 [interquartile range (IQR) 48.95-85.05] vs. 37.78 (IQR, 23.17-54.05); p < 0.001} and PWR [median, 36.89 (IQR 29.38-44.56) vs. 22.39 (IQR, 16.72-29.29); p < 0.001]. In multivariate analysis, PNR and PWR were independently associated with ruptured IA (p = 0.001 and p < 0.001, respectively). Unruptured IA subgroup analyses according to the PHASES scores showed that a higher PHASES score was associated with significantly higher NLR and erythrocyte sedimentation rate (p < 0.001 and p = 0.025) and lower PNR and PWR (p < 0.001 and p = 0.007). Conclusions We demonstrated that lower PNR and PWR levels are associated with ruptured IA and a higher PHASES score. Unlike many other inflammatory markers and bioassays, peripheral blood cell ratios are inexpensive and readily available biomarkers that may be useful for risk stratification in patients with cerebral aneurysm. However, a long-term prospective study is needed to clarify this matter.
Collapse
Affiliation(s)
- Hyun Kyung Kim
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, South Korea
| | - Kee Ook Lee
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, South Korea,*Correspondence: Kee Ook Lee ✉
| | - Seung-Hun Oh
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, South Korea
| | - Kyung-Yul Lee
- Departments of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung-Wook Choo
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, South Korea,Department of Biomedical Laboratory Science, College of Natural Science, Daejeon University, Daejeon, South Korea
| | - Ok Joon Kim
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, South Korea
| | - Tae Gon Kim
- Department of Neurosurgery, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, South Korea
| | - Sang-Heum Kim
- Department of Radiology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, South Korea
| | - Sang-Jun Na
- Department of Neurology, Konyang University College of Medicine, Daejeon, South Korea
| | - Ji Hoe Heo
- Departments of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
18
|
Li Z, Bi R, Sun S, Chen S, Chen J, Hu B, Jin H. The Role of Oxidative Stress in Acute Ischemic Stroke-Related Thrombosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8418820. [PMID: 36439687 PMCID: PMC9683973 DOI: 10.1155/2022/8418820] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/13/2022] [Accepted: 11/02/2022] [Indexed: 09/22/2023]
Abstract
Acute ischemic stroke is a serious life-threatening disease that affects almost 600 million people each year throughout the world with a mortality of more than 10%, while two-thirds of survivors remain disabled. However, the available treatments for ischemic stroke are still limited to thrombolysis and/or mechanical thrombectomy, and there is an urgent need for developing new therapeutic target. Recently, intravascular oxidative stress, derived from endothelial cells, platelets, and leukocytes, has been found to be tightly associated with stroke-related thrombosis. It not only promotes primary thrombus formation by damaging endothelial cells and platelets but also affects thrombus maturation and stability by modifying fibrin components. Thus, oxidative stress is expected to be a novel target for the prevention and treatment of ischemic stroke. In this review, we first discuss the mechanisms by which oxidative stress promotes stroke-related thrombosis, then summarize the oxidative stress biomarkers of stroke-related thrombosis, and finally put forward an antithrombotic therapy targeting oxidative stress in ischemic stroke.
Collapse
Affiliation(s)
- Zhifang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuai Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shengcai Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiefang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
19
|
Li Z, Zhang H, Baraghtha S, Mu J, Matniyaz Y, Jiang X, Wang K, Wang D, Xue YX. Short- and Mid-Term Survival Prediction in Patients with Acute Type A Aortic Dissection Undergoing Surgical Repair: Based on the Systemic Immune-Inflammation Index. J Inflamm Res 2022; 15:5785-5799. [PMID: 36238764 PMCID: PMC9553311 DOI: 10.2147/jir.s382573] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose The postoperative survival of patients with acute type A aortic dissection (aTAAD) remains unsatisfactory. The current study developed an easy-to-use survival prediction model and calculator. Methods A total of 496 patients with aTAAD undergoing surgical repair were included in this study. The systemic immune-inflammation index (SII) and other clinical features were collected and subjected to logistic and Cox regression analyses. The survival prediction model was based on Cox regression analyses and exhibited as a nomogram. For convenience of use, the nomogram was further developed into calculator software. Results We demonstrated that a higher preoperative SII was associated with in-hospital death (OR: 4.116, p < 0.001) and a higher postoperative overall survival rate (HR: 2.467, p < 0.001) in aTAAD patients undergoing surgical repair. A survival prediction model and calculator based on SII and four other clinical features were developed. The overall C-index of the model was 0.743. The areas under the curves (AUCs) of the 1- and 3-month and 1- and 3-year survival probabilities were 0.73, 0.71, 0.71 and 0.72, respectively. The model also showed good calibration and clinical utility. Conclusion Preoperative SII is significantly associated with postoperative survival. Based on SII and other clinical features, we created the first easy-to-use prediction model and calculator for predicting the postoperative survival rate in aTAAD patients, which showed good prediction performance.
Collapse
Affiliation(s)
- Zeshi Li
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Nanjing, People’s Republic of China,Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China,Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - He Zhang
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Nanjing, People’s Republic of China,Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China,Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Sulaiman Baraghtha
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China,International School, University of Mannheim, Mannheim, Baden-Württemberg, Federal Republic of Germany
| | - Jiabao Mu
- School of Data Science, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
| | - Yusanjan Matniyaz
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Xinyi Jiang
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Nanjing, People’s Republic of China,Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China,Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Kuo Wang
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China,Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of XuZhou Medical University, Nanjing, People’s Republic of China
| | - Dongjin Wang
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Nanjing, People’s Republic of China,Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China,Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China,Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of XuZhou Medical University, Nanjing, People’s Republic of China
| | - Yun Xing Xue
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China,Correspondence: Yun Xing Xue; Dongjin Wang, Email ;
| |
Collapse
|
20
|
Granja TF, Köhler D, Leiss V, Eggstein C, Nürnberg B, Rosenberger P, Beer-Hammer S. Platelets and the Cybernetic Regulation of Ischemic Inflammatory Responses through PNC Formation Regulated by Extracellular Nucleotide Metabolism and Signaling. Cells 2022; 11:cells11193009. [PMID: 36230973 PMCID: PMC9561997 DOI: 10.3390/cells11193009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Ischemic events are associated with severe inflammation and are here referred to as ischemic inflammatory response (IIR). Recent studies identified the formation of platelet–neutrophil complexes (PNC) as key players in IIR. We investigated the role of extracellular platelet nucleotide signaling in the context of IIR and defined a cybernetic circle, including description of feedback loops. Cybernetic circles seek to integrate different levels of information to understand how biological systems function. Our study specifies the components of the cybernetic system of platelets in IIR and describes the theoretical progression of IIR passing the cybernetic cycle with positive and negative feedback loops based on nucleotide-dependent signaling and functional regulation. The cybernetic components and feedback loops were explored by cytometry, immunohistological staining, functional blocking antibodies, and ADP/ATP measurements. Using several ex vivo and in vivo approaches we confirmed cybernetic parameters, such as controller, sensor, and effector (VASP phosphorylation, P2Y12, ADORAs and GPIIb/IIIa activity), as well as set points (ADP, adenosine) and interfering control and disturbance variables (ischemia). We demonstrate the impact of the regulated platelet–neutrophil complex (PNC) formation in blood and the resulting damage to the affected inflamed tissue. Taken together, extracellular nucleotide signaling, PNC formation, and tissue damage in IIR can be integrated in a controlled cybernetic circle of platelet function, as introduced through this study.
Collapse
Affiliation(s)
- Tiago F. Granja
- Lusófona’s Research Center for Biosciences & Health Technologies, CBIOS–Universidade, Campo Grande 376, 1749-024 Lisboa, Portugal
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - David Köhler
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - Veronika Leiss
- Department of Pharmacology and Experimental Therapy and Toxicology and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - Claudia Eggstein
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - Bernd Nürnberg
- Department of Pharmacology and Experimental Therapy and Toxicology and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - Sandra Beer-Hammer
- Department of Pharmacology and Experimental Therapy and Toxicology and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
- Correspondence: ; Tel.: +49-7071-29-74594
| |
Collapse
|
21
|
Sun X, Zhou M, Pu J, Wang T. Stachydrine exhibits a novel antiplatelet property and ameliorates platelet-mediated thrombo-inflammation. Biomed Pharmacother 2022; 152:113184. [PMID: 35679717 DOI: 10.1016/j.biopha.2022.113184] [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: 04/07/2022] [Revised: 05/10/2022] [Accepted: 05/22/2022] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Platelets are versatile anucleate cells involved in thrombosis as well as inflammation. Stachydrine (STA), a major bioactive compound extracted from Motherwort, has multiple pharmacological properties. Nevertheless, the significance of STA in platelet regulation and whether STA could ameliorate platelet-mediated thrombo-inflammation still remain elusive. METHODS Human platelets were used to assess the regulatory effects of STA on platelet activation and interactions with neutrophils in vitro. FeCl3 injury-induced carotid/mesenteric thrombosis and collagen/epinephrine-induced pulmonary thromboembolism model were used to explore whether STA could regulate thrombosis in vivo. Furthermore, a cecal ligation and puncture-induced sepsis model was employed to investigate the role of STA in thrombo-inflammatory diseases. RESULTS STA markedly suppressed platelet activation represented by aggregation, secretion, αIIbβ3-mediated signaling events and calcium mobilization, etc. by inhibiting agonists-induced activation signaling and potentiating cGMP-dependent inhibitory signaling. Mice receiving STA-treated platelets were less susceptible to thrombosis in vivo. In addition, decreased platelet-neutrophil interactions including platelet-neutrophil aggregates and neutrophil extracellular traps, and alleviative sepsis-induced multiorgan damage were observed due to STA-mediated platelet inhibition. CONCLUSION This study suggested the potential therapeutic role of STA in thrombotic and thrombo-inflammatory disorders.
Collapse
Affiliation(s)
- Xianting Sun
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Meng Zhou
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China.
| | - Ting Wang
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
22
|
Li X, Gao Y, Liu J, Xujian Q, Luo Q, Huang Z, Li J. Validation of Serotransferrin in the Serum as Candidate Biomarkers for the Diagnosis of Pulmonary Tuberculosis by Label-Free LC/MS. ACS OMEGA 2022; 7:24174-24183. [PMID: 35874208 PMCID: PMC9301696 DOI: 10.1021/acsomega.2c00837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
This study aimed to identify secreted protein biomarkers in serum from the label-free LC/MS proteomics of neutrophils in pulmonary tuberculosis (TB) patients for the diagnosis biomarkers of TB label-free LC/MS. The proteomic profiles of neutrophils from 15 active TB patients and 15 healthy controls (HCs) were analyzed using label-free LC/MS. We identified 358 differentially expressed proteins preliminarily, including 279 up-regulated proteins and 79 down-regulated proteins. Thirty-eight differentially expressed secreted proteins involved in the progress of platelet degranulation between TB patients and HCs were focused. Of these, serotransferrin (TRF), alpha-2-macroglobulin (AMG), alpha-1-antitrypsin (AAT), alpha-1-acid glycoprotein 1 (AAG), alpha-1-acid glycoprotein 2 (AGP2), and alpha-1B-glycoprotein (A1BG) were selected for further verification in the serum of additional 134 TB patients and 138 HCs by nephelometry and ELISA in the training set. Statistically significant differences of TRF (P < 0.0001), AAT (P < 0.0001), AAG (P < 0.0001), AGP2 (P < 0.0001), and A1BG (P = 0.0003) were observed. The serum concentration of TRF was down-regulated in TB patients compared with healthy controls, which was coincident with the proteomics results. An additional validation of TRF was performed in an independent cohort of patients with active TB (n = 46), patients with lung cancer (n = 37), 20 HCs, and patients with pneumonia (n = 35) in the test set by nephelometry. The serum expression levels of TRF in the TB patients showed lower levels compared with those in patients with pneumonia (P = 0.0125), lung cancer (P = 0.0005), HCs (P < 0.0001), and the non-TB controls (P < 0.0001). Furthermore, the AUC value of TRF was 0.647 with 90.22% sensitivity and 42.86% specificity in discriminating the TB group from the pneumonia group, 0.702 with 93.48% sensitivity and 47.16% specificity in discriminating the TB group from the lung cancer group, 0.894 with 91.30% sensitivity and 71.62% specificity in discriminating the TB group from all HCs, and 0.792 with 91.30% sensitivity and 58.90% specificity in discriminating the TB group from the non-TB controls. This study obtained the proteomic profiles of neutrophils in the TB patients and HCs, which contribute to a better understanding of the pathogenesis molecules existing in the neutrophils of pulmonary tuberculosis and provide candidate biomarkers for the diagnosis of pulmonary tuberculosis.
Collapse
Affiliation(s)
- Xue Li
- Department
of Clinical Laboratory, The First Affiliated
Hospital of Nanchang University; Institute of Infection and Immunity,
Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yujie Gao
- Department
of Clinical Laboratory, The First Affiliated
Hospital of Nanchang University; Institute of Infection and Immunity,
Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jun Liu
- Department
of Clinical Laboratory, The First Affiliated
Hospital of Nanchang University; Institute of Infection and Immunity,
Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qing Xujian
- Department
of Clinical Laboratory, The First Affiliated
Hospital of Nanchang University; Institute of Infection and Immunity,
Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qing Luo
- Department
of Clinical Laboratory, The First Affiliated
Hospital of Nanchang University; Institute of Infection and Immunity,
Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zikun Huang
- Department
of Clinical Laboratory, The First Affiliated
Hospital of Nanchang University; Institute of Infection and Immunity,
Nanchang University, Nanchang, Jiangxi 330006, China
| | - Junming Li
- Department
of Clinical Laboratory, The First Affiliated
Hospital of Nanchang University; Institute of Infection and Immunity,
Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
23
|
Zhang L, Ma J, Yang F, Li S, Ma W, Chang X, Yang L. Neuroprotective Effects of Quercetin on Ischemic Stroke: A Literature Review. Front Pharmacol 2022; 13:854249. [PMID: 35662707 PMCID: PMC9158527 DOI: 10.3389/fphar.2022.854249] [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] [Received: 01/13/2022] [Accepted: 04/28/2022] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke (IS) is characterized by high recurrence and disability; however, its therapies are very limited. As one of the effective methods of treating acute attacks of IS, intravenous thrombolysis has a clear time window. Quercetin, a flavonoid widely found in vegetables and fruits, inhibits immune cells from secreting inflammatory cytokines, thereby reducing platelet aggregation and limiting inflammatory thrombosis. In pre-clinical studies, it has been shown to exhibit neuroprotective effects in patients with ischemic brain injury. However, its specific mechanism of action remains unknown. Therefore, this review aims to use published data to elucidate the potential value of quercetin in patients with ischemic brain injury. This article also reviews the plant sources, pharmacological effects, and metabolic processes of quercetin in vivo, thus focusing on its mechanism in inhibiting immune cell activation and inflammatory thrombosis as well as promoting neuroprotection against ischemic brain injury.
Collapse
Affiliation(s)
- Leilei Zhang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Jingying Ma
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Fan Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Sishi Li
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Wangran Ma
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiang Chang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Lin Yang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
| |
Collapse
|
24
|
Zhao C, Chen Q, Li W, Zhang J, Yang C, Chen D. Multi-functional platelet membrane-camouflaged nanoparticles reduce neuronal apoptosis and regulate microglial phenotype during ischemic injury. APPLIED MATERIALS TODAY 2022; 27:101412. [DOI: 10.1016/j.apmt.2022.101412] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Affiliation(s)
- Chaoyue Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
- Changchun Children's Hospital, 1321Beian Road, Changchun, Jilin 130051, China
| | | | | | | | - Chunrong Yang
- Department of Pharmacy, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041, China
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
| |
Collapse
|
25
|
Ghasemzadeh M, Ahmadi J, Hosseini E. Platelet-leukocyte crosstalk in COVID-19: How might the reciprocal links between thrombotic events and inflammatory state affect treatment strategies and disease prognosis? Thromb Res 2022; 213:179-194. [PMID: 35397313 PMCID: PMC8969450 DOI: 10.1016/j.thromres.2022.03.022] [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] [Received: 01/22/2022] [Revised: 03/11/2022] [Accepted: 03/28/2022] [Indexed: 01/09/2023]
Abstract
Platelet-leukocyte crosstalk is commonly manifested by reciprocal links between thrombosis and inflammation. Platelet thrombus acts as a reactive matrix that recruits leukocytes to the injury site where their massive accumulation, activation and migration promote thrombotic events while triggering inflammatory responses. As a life-threatening condition with the associations between inflammation and thrombosis, COVID-19 presents diffuse alveolar damage due to exaggerated macrophage activity and cytokine storms. These events, together with direct intracellular virus invasion lead to pulmonary vascular endothelialitis, cell membranes disruption, severe endothelial injury, and thrombosis. The developing pre-alveolar thrombus provides a hyper-reactive milieu that recruits circulating leukocytes to the injury site where their activation contributes to thrombus stabilization and thrombosis propagation, primarily through the formation of Neutrophil extracellular trap (NET). NET fragments can also circulate and deposit in further distance where they may disseminate intravascular thrombosis in severe cases of disease. Thrombi may also facilitate leukocytes migration into alveoli where their accumulation and activation exacerbate cytokine storms and tissue damage, further complicating the disease. Based on these mechanisms, whether an effective anti-inflammatory protocol can prevent thrombotic events, or on the other hand; efficient antiplatelet or anticoagulant regimens may be associated with reduced cytokine storms and tissue damage, is now of interests for several ongoing researches. Thus shedding more light on platelet-leukocyte crosstalk, the review presented here discusses the detailed mechanisms by which platelets may contribute to the pathogenesis of COVID-19, especially in severe cases where their interaction with leukocytes can intensify both inflammatory state and thrombosis in a reciprocal manner.
Collapse
Affiliation(s)
- Mehran Ghasemzadeh
- Corresponding authors at: Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, Iran
| | | | - Ehteramolsadat Hosseini
- Corresponding authors at: Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, Iran
| |
Collapse
|
26
|
Yau ACY, Globisch MA, Onyeogaziri FC, Conze LL, Smith R, Jauhiainen S, Corada M, Orsenigo F, Huang H, Herre M, Olsson AK, Malinverno M, Sundell V, Rezai Jahromi B, Niemelä M, Laakso A, Garlanda C, Mantovani A, Lampugnani MG, Dejana E, Magnusson PU. Inflammation and neutrophil extracellular traps in cerebral cavernous malformation. Cell Mol Life Sci 2022; 79:206. [PMID: 35333979 PMCID: PMC8949649 DOI: 10.1007/s00018-022-04224-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023]
Abstract
Cerebral Cavernous Malformation (CCM) is a brain vascular disease with various neurological symptoms. In this study, we describe the inflammatory profile in CCM and show for the first time the formation of neutrophil extracellular traps (NETs) in rodents and humans with CCM. Through RNA-seq analysis of cerebellum endothelial cells from wild-type mice and mice with an endothelial cell-specific ablation of the Ccm3 gene (Ccm3iECKO), we show that endothelial cells from Ccm3iECKO mice have an increased expression of inflammation-related genes. These genes encode proinflammatory cytokines and chemokines, as well as adhesion molecules, which promote recruitment of inflammatory and immune cells. Similarly, immunoassays showed elevated levels of these cytokines and chemokines in the cerebellum of the Ccm3iECKO mice. Consistently, both flow cytometry and immunofluorescence analysis showed infiltration of different subsets of leukocytes into the CCM lesions. Neutrophils, which are known to fight against infection through different strategies, including the formation of NETs, represented the leukocyte subset within the most pronounced increase in CCM. Here, we detected elevated levels of NETs in the blood and the deposition of NETs in the cerebral cavernomas of Ccm3iECKO mice. Degradation of NETs by DNase I treatment improved the vascular barrier. The deposition of NETs in the cavernomas of patients with CCM confirms the clinical relevance of NETs in CCM.
Collapse
Affiliation(s)
- Anthony C Y Yau
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Maria Ascencion Globisch
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Favour Chinyere Onyeogaziri
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Lei L Conze
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Ross Smith
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Suvi Jauhiainen
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Monica Corada
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Fabrizio Orsenigo
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Hua Huang
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Melanie Herre
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Matteo Malinverno
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Veronica Sundell
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Behnam Rezai Jahromi
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Niemelä
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aki Laakso
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Maria Grazia Lampugnani
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy.,Mario Negri Institute for Pharmacological Research, 20157, Milan, Italy
| | - Elisabetta Dejana
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden.,Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden.
| |
Collapse
|
27
|
Yuan S, Chen C, Xu F, Han D, Yang R, Zheng S, Qiao M, Huang X, Lyu J. Antithrombotic Therapy Improves ICU Mortality of Septic Patients with Peripheral Vascular Disease. Int J Clin Pract 2022; 2022:1288535. [PMID: 35685503 PMCID: PMC9159174 DOI: 10.1155/2022/1288535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/03/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE The effectiveness of antithrombotic drugs for treating sepsis is controversial. Here, we explore the association between antithrombotic therapy and intensive care unit (ICU) mortality for septic patients with peripheral vascular disease. METHODS This retrospective cohort study uses data from the Medical Information Mart for Intensive Care (MIMIC)-III database. Kaplan-Meier survival analyses were used to examine mortality among different groups. Cox regression and marginal structural Cox models (MSCMs) were used to adjust for confounding factors. Main Results. The final cohort from the MIMIC-III database included 776 patients, of which 701 survived and 75 perished. The anticoagulant (AC) group and the antiplatelet-anticoagulation (AC-AP) group survived better than the group without antithrombotic treatment (non-AT). The AC and AC-AP groups showed a 0.363-fold and 0.373-fold risk of ICU mortality, respectively, compared with the non-AT group when controlling for age, gender, CRRT, alcohol, heart failure, hypertension, diabetes, obesity, renal failure, liver disease, INR, PT, PPT, and SpO2. Antiplatelet therapy did not reduce ICU mortality. The same trends were apparent from the MSCM. In addition, the AC-AP group exhibited a lower risk of bleeding complications. CONCLUSION Although the antithrombotic group (AC and AC-AP groups) demonstrated a higher sequential organ failure assessment (SOFA) score than the group without antithrombotic treatment (non-AT group), the risk of ICU mortality was lower without increasing the risk of bleeding complications. Our study further suggested that anticoagulation therapy may benefit the prognosis of septic patients with peripheral vascular disease.
Collapse
Affiliation(s)
- Shiqi Yuan
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road West, Guangzhou, Guangdong Province 510630, China
| | - Chong Chen
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, China
| | - Fengshuo Xu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road West, Guangzhou, Guangdong Province 510630, China
- School of Public Health, Xi'an Jiaotong University Health Science Center, Shaanxi Province 710061, China
| | - Didi Han
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road West, Guangzhou, Guangdong Province 510630, China
- School of Public Health, Xi'an Jiaotong University Health Science Center, Shaanxi Province 710061, China
| | - Rui Yang
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road West, Guangzhou, Guangdong Province 510630, China
- School of Public Health, Xi'an Jiaotong University Health Science Center, Shaanxi Province 710061, China
| | - Shuai Zheng
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, China
| | - Mengmeng Qiao
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, China
| | - Xiaxuan Huang
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road West, Guangzhou, Guangdong Province 510630, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road West, Guangzhou, Guangdong Province 510630, China
| |
Collapse
|
28
|
Li J, Kumari T, Barazia A, Jha V, Jeong SY, Olson A, Kim M, Lee BK, Manickam V, Song Z, Clemens R, Razani B, Kim J, Dinauer MC, Cho J. Neutrophil DREAM promotes neutrophil recruitment in vascular inflammation. J Exp Med 2022; 219:e20211083. [PMID: 34751735 PMCID: PMC8719643 DOI: 10.1084/jem.20211083] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/21/2021] [Accepted: 10/19/2021] [Indexed: 01/02/2023] Open
Abstract
The interaction between neutrophils and endothelial cells is critical for the pathogenesis of vascular inflammation. However, the regulation of neutrophil adhesive function remains not fully understood. Intravital microscopy demonstrates that neutrophil DREAM promotes neutrophil recruitment to sites of inflammation induced by TNF-α but not MIP-2 or fMLP. We observe that neutrophil DREAM represses expression of A20, a negative regulator of NF-κB activity, and enhances expression of pro-inflammatory molecules and phosphorylation of IκB kinase (IKK) after TNF-α stimulation. Studies using genetic and pharmacologic approaches reveal that DREAM deficiency and IKKβ inhibition significantly diminish the ligand-binding activity of β2 integrins in TNF-α-stimulated neutrophils or neutrophil-like HL-60 cells. Neutrophil DREAM promotes degranulation through IKKβ-mediated SNAP-23 phosphorylation. Using sickle cell disease mice lacking DREAM, we show that hematopoietic DREAM promotes vaso-occlusive events in microvessels following TNF-α challenge. Our study provides evidence that targeting DREAM might be a novel therapeutic strategy to reduce excessive neutrophil recruitment in inflammatory diseases.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL
| | - Tripti Kumari
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Andrew Barazia
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL
| | - Vishwanath Jha
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Si-Yeon Jeong
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL
| | - Amber Olson
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Mijeong Kim
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX
| | - Bum-Kyu Lee
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX
| | - Vijayprakash Manickam
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Zhimin Song
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Regina Clemens
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Babak Razani
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO
- John Cochran VA Medical Center, St. Louis, MO
| | - Jonghwan Kim
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX
| | - Mary C. Dinauer
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
29
|
Banka AL, Eniola-Adefeso O. Method article: an in vitro blood flow model to advance the study of platelet adhesion utilizing a damaged endothelium. Platelets 2021; 33:692-699. [PMID: 34927530 DOI: 10.1080/09537104.2021.1988550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In vitro flow assays utilizing microfluidic devices are often used to study human platelets as an alternative to the costly animal models of hemostasis and thrombosis that may not accurately represent human platelet behavior in vivo. Here, we present a tunable in vitro model to study platelet behavior in human whole blood flow that includes both an inflamed, damaged endothelium and exposed extracellular matrix. We demonstrate that the model is adaptable across various anticoagulants, shear rates, and proteins for endothelial cell culture without the need for a complicated, custom-designed device. Furthermore, we verified the ability of this 'damaged endothelium' model as a screening method for potential anti-platelet or anti-thrombotic compounds using a P2Y12 receptor antagonist (ticagrelor), a pan-selectin inhibitor (Bimosiamose), and a histamine receptor antagonist (Cimetidine). These compounds significantly decreased platelet adhesion to the damaged endothelium, highlighting that this model can successfully screen anti-platelet compounds that target platelets directly or the endothelium indirectly.
Collapse
Affiliation(s)
- Alison Leigh Banka
- Department of Chemical Engineering, University of Michigan, Ann Arbor, USA
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
| |
Collapse
|
30
|
Systemic inflammation as a risk factor for portal vein thrombosis in cirrhosis: a prospective longitudinal study. Eur J Gastroenterol Hepatol 2021; 33:e108-e113. [PMID: 33208682 DOI: 10.1097/meg.0000000000001982] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND AND AIMS Various risk factors for portal vein thrombosis (PVT) development in patients with cirrhosis have been identified, but the role of systemic inflammatory reaction is unknown. The study aims to assess the association between markers of systemic inflammation and PVT in cirrhosis. METHODS Between January 2014 and October 2015, 107 outpatients with cirrhosis and no PVT were recruited, and followed till February 2017. White blood cell count, serum concentrations of high-sensitive C-reactive protein, ferritin, tumor necrosis factor-alpha and interleukin-6 (IL-6) were evaluated at baseline and every 3 or 6 months till PVT diagnosis or end of follow-up. RESULTS Median age, model for end-stage liver disease (MELD) score and follow-up period of the studied population was 55 years (IQR 46-62 years), 9.6 points (IQR 7.5-12 points) and 19 months (12-24 months), respectively. PVT developed in 10.3% of the patients. Lymphocyte count below 1.2 ´ 109/L [hazard ratio, 6.04; 95% confidence interval (CI), 1.29-28.2; P = 0.022], IL-6 above 5.5 pg/mL (hazard ratio, 5.64; 95% CI, 1.21-26.33; P = 0.028) and neutrophil-to-lymphocyte ratio (hazard ratio, 1.46; 95% CI, 1.04-2.04; P = 0.028) were associated with a higher risk of PVT development. IL-6 and lymphopenia remained associated with subsequent PVT development after adjustment for nonselective beta-blockers, spleen size, portosystemic collaterals, oesophageal varices (grade ≥2) and ascites, but also with alcohol as the cause for cirrhosis and MELD ≥13. CONCLUSION In patients with cirrhosis, markers of systemic inflammation IL-6 and lymphopenia are predictive of PVT independently of markers of portal hypertension. These results draw our attention on a factor so far overlooked in the pathogenesis of PVT.
Collapse
|
31
|
Kim SJ, Carestia A, McDonald B, Zucoloto AZ, Grosjean H, Davis RP, Turk M, Naumenko V, Antoniak S, Mackman N, Abdul-Cader MS, Abdul-Careem MF, Hollenberg MD, Jenne CN. Platelet-Mediated NET Release Amplifies Coagulopathy and Drives Lung Pathology During Severe Influenza Infection. Front Immunol 2021; 12:772859. [PMID: 34858432 PMCID: PMC8632260 DOI: 10.3389/fimmu.2021.772859] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 01/30/2023] Open
Abstract
The influenza A virus (IAV) causes a respiratory tract infection with approximately 10% of the population infected by the virus each year. Severe IAV infection is characterized by excessive inflammation and tissue pathology in the lungs. Platelet and neutrophil recruitment to the lung are involved in the pathogenesis of IAV, but the specific mechanisms involved have not been clarified. Using confocal intravital microscopy in a mouse model of IAV infection, we observed profound neutrophil recruitment, platelet aggregation, neutrophil extracellular trap (NET) production and thrombin activation within the lung microvasculature in vivo. Importantly, deficiency or antagonism of the protease-activated receptor 4 (PAR4) reduced platelet aggregation, NET production, and neutrophil recruitment. Critically, inhibition of thrombin or PAR4 protected mice from virus-induced lung tissue damage and edema. Together, these data imply thrombin-stimulated platelets play a critical role in the activation/recruitment of neutrophils, NET release and directly contribute to IAV pathogenesis in the lung.
Collapse
MESH Headings
- Animals
- Blood Coagulation Disorders/immunology
- Blood Coagulation Disorders/metabolism
- Blood Coagulation Disorders/virology
- Blood Platelets/immunology
- Blood Platelets/metabolism
- Blood Platelets/virology
- Disease Models, Animal
- Extracellular Traps/immunology
- Extracellular Traps/metabolism
- Extracellular Traps/virology
- Female
- Humans
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/physiology
- Influenza, Human/immunology
- Influenza, Human/metabolism
- Influenza, Human/virology
- Lung/immunology
- Lung/metabolism
- Lung/virology
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Confocal
- Neutrophil Infiltration/immunology
- Neutrophils/immunology
- Neutrophils/metabolism
- Neutrophils/virology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/metabolism
- Orthomyxoviridae Infections/virology
- Platelet Aggregation/immunology
- Mice
Collapse
Affiliation(s)
- Seok-Joo Kim
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Agostina Carestia
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Braedon McDonald
- Department of Critical Care Medicine, University of Calgary, Calgary, AB, Canada
| | - Amanda Z. Zucoloto
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Heidi Grosjean
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Rachelle P. Davis
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Madison Turk
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Victor Naumenko
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Silvio Antoniak
- UNC Blood Research Center, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nigel Mackman
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | | | - Morley D. Hollenberg
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Craig N. Jenne
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
32
|
Jha V, Kumari T, Manickam V, Assar Z, Olson KL, Min JK, Cho J. ERO1-PDI Redox Signaling in Health and Disease. Antioxid Redox Signal 2021; 35:1093-1115. [PMID: 34074138 PMCID: PMC8817699 DOI: 10.1089/ars.2021.0018] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Significance: Protein disulfide isomerase (PDI) and endoplasmic reticulum oxidoreductase 1 (ERO1) are crucial for oxidative protein folding in the endoplasmic reticulum (ER). These enzymes are frequently overexpressed and secreted, and they contribute to the pathology of neurodegenerative, cardiovascular, and metabolic diseases. Recent Advances: Tissue-specific knockout mouse models and pharmacologic inhibitors have been developed to advance our understanding of the cell-specific functions of PDI and ERO1. In addition to their roles in protecting cells from the unfolded protein response and oxidative stress, recent studies have revealed that PDI and ERO1 also function outside of the cells. Critical Issues: Despite the well-known contributions of PDI and ERO1 to specific disease pathology, the detailed molecular and cellular mechanisms underlying these activities remain to be elucidated. Further, although PDI and ERO1 inhibitors have been identified, the results from previous studies require careful evaluation, as many of these agents are not selective and may have significant cytotoxicity. Future Directions: The functions of PDI and ERO1 in the ER have been extensively studied. Additional studies will be required to define their functions outside the ER.
Collapse
Affiliation(s)
- Vishwanath Jha
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tripti Kumari
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Vijayprakash Manickam
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zahra Assar
- Cayman Chemical Company, Inc., Ann Arbor, Michigan, USA
| | - Kirk L Olson
- Cayman Chemical Company, Inc., Ann Arbor, Michigan, USA
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
33
|
Chu SJ, Tang SE, Pao HP, Wu SY, Liao WI. Protease-Activated Receptor-1 Antagonist Protects Against Lung Ischemia/Reperfusion Injury. Front Pharmacol 2021; 12:752507. [PMID: 34658893 PMCID: PMC8514687 DOI: 10.3389/fphar.2021.752507] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/20/2021] [Indexed: 01/14/2023] Open
Abstract
Protease-activated receptor (PAR)-1 is a thrombin-activated receptor that plays an essential role in ischemia/reperfusion (IR)-induced acute inflammation. PAR-1 antagonists have been shown to alleviate injuries in various IR models. However, the effect of PAR-1 antagonists on IR-induced acute lung injury (ALI) has not yet been elucidated. This study aimed to investigate whether PAR-1 inhibition could attenuate lung IR injury. Lung IR was induced in an isolated perfused rat lung model. Male rats were treated with the specific PAR-1 antagonist SCH530348 (vorapaxar) or vehicle, followed by ischemia for 40 min and reperfusion for 60 min. To examine the role of PAR-1 and the mechanism of SCH530348 in lung IR injury, western blotting and immunohistochemical analysis of lung tissue were performed. In vitro, mouse lung epithelial cells (MLE-12) were treated with SCH530348 or vehicle and subjected to hypoxia-reoxygenation (HR). We found that SCH530348 decreased lung edema and neutrophil infiltration, attenuated thrombin production, reduced inflammatory factors, including cytokine-induced neutrophil chemoattractant-1, interleukin-6 and tumor necrosis factor-α, mitigated lung cell apoptosis, and downregulated the phosphoinositide 3-kinase (PI3K), nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways in IR-injured lungs. In addition, SCH530348 prevented HR-induced NF-κB activation and inflammatory chemokine production in MLE12 cells. Our results demonstrate that SCH530348 exerts protective effects by blocking PAR-1 expression and modulating the downstream PI3K, NF-κB and MAPK pathways. These findings indicate that the PAR-1 antagonist protects against IR-induced ALI and is a potential therapeutic candidate for lung protection following IR injury.
Collapse
Affiliation(s)
- Shi-Jye Chu
- Department of Internal Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Shih-En Tang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan.,Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| |
Collapse
|
34
|
Chebbo M, Duez C, Alessi MC, Chanez P, Gras D. Platelets: a potential role in chronic respiratory diseases? Eur Respir Rev 2021; 30:30/161/210062. [PMID: 34526315 PMCID: PMC9488457 DOI: 10.1183/16000617.0062-2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/05/2021] [Indexed: 12/21/2022] Open
Abstract
Platelets are small anucleate cells known for their role in haemostasis and thrombosis. In recent years, an increasing number of observations have suggested that platelets are also immune cells and key modulators of immunity. They express different receptors and molecules that allow them to respond to pathogens, and to interact with other immune cells. Platelets were linked to the pathogenesis of some inflammatory disorders including respiratory diseases such as asthma and idiopathic pulmonary fibrosis. Here, we discuss the involvement of platelets in different immune responses, and we focus on their potential role in various chronic lung diseases. In addition to their essential role in haemostasis and thrombosis, platelets are strong modulators of different immune responses, and could be involved in the physiopathology of several chronic airway diseaseshttps://bit.ly/3cB6Xnj
Collapse
Affiliation(s)
| | | | - Marie C Alessi
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France.,APHM, CHU de la Timone, Laboratoire d'hématologie, Marseille, France
| | - Pascal Chanez
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France.,APHM, Hôpital NORD, Clinique des Bronches, Allergie et Sommeil, Marseille, France
| | - Delphine Gras
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France
| |
Collapse
|
35
|
Bi R, Chen S, Chen S, Peng Q, Jin H, Hu B. The role of leukocytes in acute ischemic stroke-related thrombosis: a notable but neglected topic. Cell Mol Life Sci 2021; 78:6251-6264. [PMID: 34398251 PMCID: PMC11072166 DOI: 10.1007/s00018-021-03897-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/15/2021] [Accepted: 07/02/2021] [Indexed: 12/19/2022]
Abstract
Ischemic stroke is one of the most serious diseases today, and only a minority of patients are provided with effective clinical treatment. Importantly, leukocytes have gradually been discovered to play vital roles in stroke thrombosis, including promoting the activation of thrombin and the adhesion and aggregation of platelets. However, they have not received enough attention in the field of acute ischemic stroke. It is possible that we could not only prevent stroke-related thrombosis by inhibiting leukocyte activation, but also target leukocyte components to dissolve thrombi in the cerebral artery. In this review, we expound the mechanisms by which leukocytes are activated and participate in the formation of stroke thrombus, then describe the histopathology of leukocytes in thrombi of stroke patients and the influence of leukocyte composition on vascular recanalization effects and patient prognosis. Finally, we discuss the relevant antithrombotic strategies targeting leukocytes.
Collapse
Affiliation(s)
- Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Shengcai Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Shaolin Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Qiwei Peng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
36
|
Hemogram-derived ratios as prognostic markers of ICU admission in COVID-19. BMC Emerg Med 2021; 21:89. [PMID: 34315437 PMCID: PMC8314257 DOI: 10.1186/s12873-021-00480-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/04/2021] [Indexed: 01/08/2023] Open
Abstract
Background The vast impact of COVID-19 call for the identification of clinical parameter that can help predict a torpid evolution. Among these, endothelial injury has been proposed as one of the main pathophysiological mechanisms underlying the disease, promoting a hyperinflammatory and prothrombotic state leading to worse clinical outcomes. Leukocytes and platelets play a key role in inflammation and thrombogenesis, hence the objective of the current study was to study whether neutrophil-to-lymphocyte ratio (NLR), platelets-to-lymphocyte ratio (PLR), the systemic immune-inflammation index (SII) as well as the new parameter neutrophil-to-platelet ratio (NPR), could help identify patients who at risk of admission at Intensive Care Units. Methods A retrospective observational study was performed at HM Hospitales including electronic health records from 2245 patients admitted due to COVID-19 from March 1 to June 10, 2020. Patients were divided into two groups, admitted at ICU or not. Results Patients who were admitted at the ICU had significantly higher values in all hemogram-derived ratios at the moment of hospital admission compared to those who did not need ICU admission. Specifically, we found significant differences in NLR (6.9 [4–11.7] vs 4.1 [2.6–7.6], p < 0.0001), PLR (2 [1.4–3.3] vs 1.9 [1.3–2.9], p = 0.023), NPR (3 [2.1–4.2] vs 2.3 [1.6–3.2], p < 0.0001) and SII (13 [6.5–25.7] vs 9 [4.9–17.5], p < 0.0001) compared to those who did not require ICU admission. After multivariable logistic regression models, NPR was the hemogram-derived ratio with the highest predictive value of ICU admission, (OR 1.11 (95% CI: 0.98–1.22, p = 0.055). Conclusions Simple, hemogram-derived ratios obtained from early hemogram at hospital admission, especially the novelty NPR, have shown to be useful predictors of risk of ICU admission in patients hospitalized due to COVID-19. Supplementary Information The online version contains supplementary material available at 10.1186/s12873-021-00480-w.
Collapse
|
37
|
Gil-Etayo FJ, Garcinuño S, Lalueza A, Díaz-Simón R, García-Reyne A, Pleguezuelo DE, Cabrera-Marante O, Rodriguez-Frias EA, Perez-Rivilla A, Serrano M, Serrano A. Anti-Phospholipid Antibodies and COVID-19 Thrombosis: A Co-Star, Not a Supporting Actor. Biomedicines 2021; 9:biomedicines9080899. [PMID: 34440103 PMCID: PMC8389622 DOI: 10.3390/biomedicines9080899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Background: COVID-19 clinical features include a hypercoagulable state that resembles the antiphospholipid syndrome (APS), a disease characterized by thrombosis and presence of antiphospholipid antibodies (aPL). The relationship between aPL-presence and the appearance of thrombi as well as the transience or permanence of aPL in COVID-19 patients is not sufficiently clear. Methods: A group of 360 COVID-19 patients were followed-up for 6 months. Classic aPL, anti-B2GPI IgA, anti-phosphatidylserine/prothrombin IgG/M and anti-SARS-CoV-2 antibodies were determined at acute phase and >12 weeks later. The reference group included 143 healthy volunteers of the same age-range distribution. Results: aPL prevalence was similar in COVID-19 patients and the reference population. aPL presence in both determinations was significantly associated with thrombosis (OR: 2.33 and 3.71), strong agreement being found for classic aPL and anti-B2GPI IgA (Weighted kappa: 0.85–0.91). Thrombosis-associated aPL occurred a median of 17 days after hospital admission (IQR: 6–28) vs. 4 days for the rest (IQR: 3–7). Although anti-SARS-CoV-2 antibodies levels increased during convalescence, aPL hardly changed. Conclusions: Most COVID-19 patients would carry these aPL before the infection. At least two mechanisms could be behind thrombosis, early immune-dysregulation-mediated thrombosis after infection and belated-aPL-mediated thrombosis, with SARS-CoV-2 behaving as a second hit.
Collapse
Affiliation(s)
- Francisco Javier Gil-Etayo
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (F.J.G.-E.); (D.E.P.); (O.C.-M.); (E.A.R.-F.)
| | - Sara Garcinuño
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
| | - Antonio Lalueza
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (A.L.); (R.D.-S.); (A.G.-R.)
| | - Raquel Díaz-Simón
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (A.L.); (R.D.-S.); (A.G.-R.)
| | - Ana García-Reyne
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (A.L.); (R.D.-S.); (A.G.-R.)
| | - Daniel Enrique Pleguezuelo
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (F.J.G.-E.); (D.E.P.); (O.C.-M.); (E.A.R.-F.)
| | - Oscar Cabrera-Marante
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (F.J.G.-E.); (D.E.P.); (O.C.-M.); (E.A.R.-F.)
| | - Edgard Alfonso Rodriguez-Frias
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (F.J.G.-E.); (D.E.P.); (O.C.-M.); (E.A.R.-F.)
| | - Alfredo Perez-Rivilla
- Department of Microbiology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | - Manuel Serrano
- Department of Immunology, Hospital Universitario Clínico San Carlos, 28041 Madrid, Spain;
| | - Antonio Serrano
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (F.J.G.-E.); (D.E.P.); (O.C.-M.); (E.A.R.-F.)
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
- Department of Epidemiology, Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-659-496-544
| |
Collapse
|
38
|
López-Escobar A, Madurga R, Castellano JM, Ruiz de Aguiar S, Velázquez S, Bucar M, Jimeno S, Ventura PS. Hemogram as marker of in-hospital mortality in COVID-19. J Investig Med 2021; 69:962-969. [PMID: 33849952 PMCID: PMC8050870 DOI: 10.1136/jim-2021-001810] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 01/08/2023]
Abstract
The clinical impact of COVID-19 disease calls for the identification of routine variables to identify patients at increased risk of death. Current understanding of moderate-to-severe COVID-19 pathophysiology points toward an underlying cytokine release driving a hyperinflammatory and procoagulant state. In this scenario, white blood cells and platelets play a direct role as effectors of such inflammation and thrombotic response. We investigate whether hemogram-derived ratios such as neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio and the systemic immune-inflammation index may help to identify patients at risk of fatal outcomes. Activated platelets and neutrophils may be playing a decisive role during the thromboinflammatory phase of COVID-19 so, in addition, we introduce and validate a novel marker, the neutrophil-to-platelet ratio (NPR).Two thousand and eighty-eight hospitalized patients with COVID-19 admitted at any of the hospitals of HM Hospitales group in Spain, from March 1 to June 10, 2020, were categorized according to the primary outcome of in-hospital death.Baseline values, as well as the rate of increase of the four ratios analyzed were significantly higher at hospital admission in patients who died than in those who were discharged (p<0.0001). In multivariable logistic regression models, NLR (OR 1.05; 95% CI 1.02 to 1.08, p=0.00035) and NPR (OR 1.23; 95% CI 1.12 to 1.36, p<0.0001) were significantly and independently associated with in-hospital mortality.According to our results, hemogram-derived ratios obtained at hospital admission, as well as the rate of change during hospitalization, may easily detect, primarily using NLR and the novel NPR, patients with COVID-19 at high risk of in-hospital mortality.
Collapse
Affiliation(s)
- Alejandro López-Escobar
- Pediatrics Department, HM Hospitales, Madrid, Spain
- Faculty of Medicine, Universidad San Pablo CEU, Madrid, Spain
- Fundación de Investigación, HM Hospitales, Madrid, Spain
| | - Rodrigo Madurga
- Fundación de Investigación, HM Hospitales, Madrid, Spain
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Comunidad de Madrid, Spain
| | - José María Castellano
- Faculty of Medicine, Universidad San Pablo CEU, Madrid, Spain
- Fundación de Investigación, HM Hospitales, Madrid, Spain
- Cardiology Department, Hospital Universitario HM Montepríncipe, HM Hospitales, Madrid, Spain
| | - Santiago Ruiz de Aguiar
- Fundación de Investigación, HM Hospitales, Madrid, Spain
- Medical Management, HM Hospitales, Madrid, Spain
| | - Sara Velázquez
- Fundación de Investigación, HM Hospitales, Madrid, Spain
- Anaesthesia Department, HM Hospitales, Madrid, Spain
- Anaesthesia Department, Hospital Universitario Santa Cristina, Madrid, Spain
| | - Marina Bucar
- Fundación de Investigación, HM Hospitales, Madrid, Spain
- Internal Medicine Department, HM Hospitales, Madrid, Spain
| | - Sara Jimeno
- Pediatrics Department, HM Hospitales, Madrid, Spain
- Faculty of Medicine, Universidad San Pablo CEU, Madrid, Spain
- Fundación de Investigación, HM Hospitales, Madrid, Spain
| | - Paula Sol Ventura
- Fundación de Investigación, HM Hospitales, Madrid, Spain
- Pediatrics Department Hospital Universitario HM Nens, HM Hospitales, Barcelona, Madrid, Spain
| |
Collapse
|
39
|
Bartig KA, Lee KE, Mosher DF, Mathur SK, Johansson MW. Platelet association with leukocytes in active eosinophilic esophagitis. PLoS One 2021; 16:e0250521. [PMID: 33891621 PMCID: PMC8064567 DOI: 10.1371/journal.pone.0250521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/07/2021] [Indexed: 12/27/2022] Open
Abstract
We previously demonstrated that the percentage of blood eosinophils that are associated with platelets and thus positive for CD41 (integrin αIIb-subunit) correlates with and predicts peak eosinophil count (PEC) in biopsies of eosinophilic esophagitis (EoE) patients after treatment. Thus, flow cytometric determination of CD41+ eosinophils is a potential measure of EoE disease activity. Determinants of association of platelets with eosinophils and other leukocytes in EoE are largely unknown. The objectives of this study were to test the hypotheses that platelets associate with blood leukocytes other than eosinophils in EoE and that such associations also predict EoE activity. Whole blood flow cytometry was performed on samples from 25 subjects before and after two months of standard of care EoE treatment. CD41 positivity of cells within gates for eosinophils, neutrophils, monocytes, lymphocytes, and natural killer cells was compared. We found that percent CD41+ neutrophils, monocytes, and eosinophils correlated with one another such that principal component analysis of the five cell types identified “myeloid” and “lymphoid” factors. Percent CD41+ neutrophils or monocytes, or the myeloid factor, like CD41+ eosinophils, correlated with PEC after treatment, and CD41+ neutrophils or the myeloid factor predicted PEC < 6/high power field after treatment, albeit with lower area under the curve than for CD41+ eosinophils. We conclude that the processes driving platelets to associate with eosinophils in EoE also drive association of platelets with neutrophils and monocytes and that association of platelets with all three cell types is related to disease activity. Clinicaltrials.gov identifier: NCT02775045.
Collapse
Affiliation(s)
- Kelly A. Bartig
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Kristine E. Lee
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Deane F. Mosher
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Sameer K. Mathur
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Mats W. Johansson
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
40
|
Blanch-Ruiz MA, Ortega-Luna R, Martínez-Cuesta MÁ, Álvarez Á. The Neutrophil Secretome as a Crucial Link between Inflammation and Thrombosis. Int J Mol Sci 2021; 22:4170. [PMID: 33920656 PMCID: PMC8073391 DOI: 10.3390/ijms22084170] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases are a leading cause of death. Blood-cell interactions and endothelial dysfunction are fundamental in thrombus formation, and so further knowledge of the pathways involved in such cellular crosstalk could lead to new therapeutical approaches. Neutrophils are secretory cells that release well-known soluble inflammatory signaling mediators and other complex cellular structures whose role is not fully understood. Studies have reported that neutrophil extracellular vesicles (EVs) and neutrophil extracellular traps (NETs) contribute to thrombosis. The objective of this review is to study the role of EVs and NETs as key factors in the transition from inflammation to thrombosis. The neutrophil secretome can promote thrombosis due to the presence of different factors in the EVs bilayer that can trigger blood clotting, and to the release of soluble mediators that induce platelet activation or aggregation. On the other hand, one of the main pathways by which NETs induce thrombosis is through the creation of a scaffold to which platelets and other blood cells adhere. In this context, platelet activation has been associated with the induction of NETs release. Hence, the structure and composition of EVs and NETs, as well as the feedback mechanism between the two processes that causes pathological thrombus formation, require exhaustive analysis to clarify their role in thrombosis.
Collapse
Affiliation(s)
- María Amparo Blanch-Ruiz
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
| | - Raquel Ortega-Luna
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
| | - María Ángeles Martínez-Cuesta
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| | - Ángeles Álvarez
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| |
Collapse
|
41
|
Ansari J, Senchenkova EY, Vital SA, Al-Yafeai Z, Kaur G, Sparkenbaugh EM, Orr AW, Pawlinski R, Hebbel RP, Granger DN, Kubes P, Gavins FNE. Targeting the AnxA1/Fpr2/ALX pathway regulates neutrophil function, promoting thromboinflammation resolution in sickle cell disease. Blood 2021; 137:1538-1549. [PMID: 33512489 PMCID: PMC7976506 DOI: 10.1182/blood.2020009166] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/28/2020] [Indexed: 12/19/2022] Open
Abstract
Neutrophils play a crucial role in the intertwined processes of thrombosis and inflammation. An altered neutrophil phenotype may contribute to inadequate resolution, which is known to be a major pathophysiological contributor of thromboinflammatory conditions such as sickle cell disease (SCD). The endogenous protein annexin A1 (AnxA1) facilitates inflammation resolution via formyl peptide receptors (FPRs). We sought to comprehensively elucidate the functional significance of targeting the neutrophil-dependent AnxA1/FPR2/ALX pathway in SCD. Administration of AnxA1 mimetic peptide AnxA1Ac2-26 ameliorated cerebral thrombotic responses in Sickle transgenic mice via regulation of the FPR2/ALX (a fundamental receptor involved in resolution) pathway. We found direct evidence that neutrophils with SCD phenotype play a key role in contributing to thromboinflammation. In addition, AnxA1Ac2-26 regulated activated SCD neutrophils through protein kinase B (Akt) and extracellular signal-regulated kinases (ERK1/2) to enable resolution. We present compelling conceptual evidence that targeting the AnxA1/FPR2/ALX pathway may provide new therapeutic possibilities against thromboinflammatory conditions such as SCD.
Collapse
Affiliation(s)
- Junaid Ansari
- Department of Molecular and Cellular Physiology
- Department of Neurology, and
| | | | | | - Zaki Al-Yafeai
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | | | - Erica M Sparkenbaugh
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - A Wayne Orr
- Department of Molecular and Cellular Physiology
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - Rafal Pawlinski
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Robert P Hebbel
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | | | - Paul Kubes
- Departments of Physiology and Pharmacology, Microbiology and Immunology and Critical Care Medicine, Snyder Institute for Chronic Disease, University of Calgary, Calgary, AB, Canada; and
| | - Felicity N E Gavins
- Department of Molecular and Cellular Physiology
- Department of Neurology, and
- Department of Life Sciences, Brunel University London, United Kingdom
| |
Collapse
|
42
|
Robba C, Battaglini D, Ball L, Valbusa A, Porto I, Della Bona R, La Malfa G, Patroniti N, Brunetti I, Loconte M, Bassetti M, Giacobbe DR, Vena A, Silva CLM, Rocco PRM, Pelosi P. Coagulative Disorders in Critically Ill COVID-19 Patients with Acute Distress Respiratory Syndrome: A Critical Review. J Clin Med 2021; 10:E140. [PMID: 33401632 PMCID: PMC7795033 DOI: 10.3390/jcm10010140] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
In critically ill patients with acute respiratory distress syndrome (ARDS) coronavirus disease 2019 (COVID-19), a high incidence of thromboembolic and hemorrhagic events is reported. COVID-19 may lead to impairment of the coagulation cascade, with an imbalance in platelet function and the regulatory mechanisms of coagulation and fibrinolysis. Clinical manifestations vary from a rise in laboratory markers and subclinical microthrombi to thromboembolic events, bleeding, and disseminated intravascular coagulation. After an inflammatory trigger, the mechanism for activation of the coagulation cascade in COVID-19 is the tissue factor pathway, which causes endotoxin and tumor necrosis factor-mediated production of interleukins and platelet activation. The consequent massive infiltration of activated platelets may be responsible for inflammatory infiltrates in the endothelial space, as well as thrombocytopenia. The variety of clinical presentations of the coagulopathy confronts the clinician with the difficult questions of whether and how to provide optimal supportive care. In addition to coagulation tests, advanced laboratory tests such as protein C, protein S, antithrombin, tissue factor pathway inhibitors, D-dimers, activated factor Xa, and quantification of specific coagulation factors can be useful, as can thromboelastography or thromboelastometry. Treatment should be tailored, focusing on the estimated risk of bleeding and thrombosis. The aim of this review is to explore the pathophysiology and clinical evidence of coagulation disorders in severe ARDS-related COVID-19 patients.
Collapse
Affiliation(s)
- Chiara Robba
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Denise Battaglini
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
| | - Lorenzo Ball
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Alberto Valbusa
- Dipartimento CardioToracoVascolare, Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy; (A.V.); (I.P.); (R.D.B.); (G.L.M.)
| | - Italo Porto
- Dipartimento CardioToracoVascolare, Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy; (A.V.); (I.P.); (R.D.B.); (G.L.M.)
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Roberta Della Bona
- Dipartimento CardioToracoVascolare, Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy; (A.V.); (I.P.); (R.D.B.); (G.L.M.)
| | - Giovanni La Malfa
- Dipartimento CardioToracoVascolare, Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy; (A.V.); (I.P.); (R.D.B.); (G.L.M.)
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Nicolò Patroniti
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Iole Brunetti
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
| | - Maurizio Loconte
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
| | - Matteo Bassetti
- Infectious Diseases Unit, Ospedale Policlinico San Martino, IRCCS, 16132 Genoa, Italy; (M.B.); (D.R.G.); (A.V.)
- Department of Health Sciences (DISSAL), University of Genoa, 16132 Genoa, Italy
| | - Daniele R. Giacobbe
- Infectious Diseases Unit, Ospedale Policlinico San Martino, IRCCS, 16132 Genoa, Italy; (M.B.); (D.R.G.); (A.V.)
| | - Antonio Vena
- Infectious Diseases Unit, Ospedale Policlinico San Martino, IRCCS, 16132 Genoa, Italy; (M.B.); (D.R.G.); (A.V.)
| | - Claudia Lucia M. Silva
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil;
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil;
| | - Paolo Pelosi
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| |
Collapse
|
43
|
Vital SA, Senchenkova EY, Ansari J, Gavins FNE. Targeting AnxA1/Formyl Peptide Receptor 2 Pathway Affords Protection against Pathological Thrombo-Inflammation. Cells 2020; 9:cells9112473. [PMID: 33202930 PMCID: PMC7697101 DOI: 10.3390/cells9112473] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke is a leading cause of death and disability globally and is associated with a number of co-morbidities including sepsis and sickle cell disease (SCD). Despite thrombo-inflammation underlying these co-morbidities, its pathogenesis remains complicated and drug discovery programs aimed at reducing and resolving the detrimental effects remain a major therapeutic challenge. The objective of this study was to assess whether the anti-inflammatory pro-resolving protein Annexin A1 (AnxA1) was able to reduce inflammation-induced thrombosis and suppress platelet activation and thrombus formation in the cerebral microvasculature. Using two distinct models of pathological thrombo-inflammation (lipopolysaccharide (LPS) and sickle transgenic mice (STM)), thrombosis was induced in the murine brain using photoactivation (light/dye) coupled with intravital microscopy. The heightened inflammation-induced microvascular thrombosis present in these two distinct thrombo-inflammatory models was inhibited significantly by the administration of AnxA1 mimetic peptide AnxA1Ac2-26 (an effect more pronounced in the SCD model vs. the endotoxin model) and mediated by the key resolution receptor, Fpr2/ALX. Furthermore, AnxA1Ac2-26 treatment was able to hamper platelet aggregation by reducing platelet stimulation and aggregation (by moderating αIIbβ3 and P-selectin). These findings suggest that targeting the AnxA1/Fpr2/ALX pathway represents an attractive novel treatment strategy for resolving thrombo-inflammation, counteracting e.g., stroke in high-risk patient cohorts.
Collapse
Affiliation(s)
- Shantel A. Vital
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
| | - Elena Y. Senchenkova
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
| | - Junaid Ansari
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA
| | - Felicity N. E. Gavins
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
- Correspondence: ; Tel.: +44-(0)-1895-267151
| |
Collapse
|
44
|
Li J, Jeong SY, Xiong B, Tseng A, Mahon AB, Isaacman S, Gordeuk VR, Cho J. Repurposing pyridoxamine for therapeutic intervention of intravascular cell-cell interactions in mouse models of sickle cell disease. Haematologica 2020; 105:2407-2419. [PMID: 33054081 PMCID: PMC7556679 DOI: 10.3324/haematol.2019.226720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/29/2019] [Indexed: 12/25/2022] Open
Abstract
Adherent neutrophils on vascular endothelium positively contribute to cell-cell aggregation and vaso-occlusion in sickle cell disease. In the present study, we demonstrated that pyridoxamine, a derivative of vitamin B6, might be a therapeutic agent to alleviate intravascular cell-cell aggregation in sickle cell disease. Using real-time intravital microscopy, we found that one oral administration of pyridoxamine dose-dependently increased the rolling influx of neutrophils and reduced neutrophil adhesion to endothelial cells in cremaster microvessels of sickle cell disease mice challenged with hypoxia-reoxygenation. Short-term treatment also mitigated neutrophil-endothelial cell and neutrophil-platelet interactions in the microvessels and improved the survival of sickle cell disease mice challenged with tumor necrosis factor-α. The inhibitory effects of pyridoxamine on intravascular cell-cell interactions were potentiated by co-treatment with hydroxyurea. We observed that long-term (5.5 months) oral treatment with pyridoxamine significantly diminished the adhesive function of neutrophils and platelets and down-regulated the expression of E-selectin and intercellular adhesion molecule-1 on the vascular endothelium in tumor necrosis factor-α-challenged sickle cell disease mice. Ex vivo studies revealed that the surface amount of αMβ2 integrin was significantly decreased in stimulated neutrophils isolated from sickle cell disease mice treated with pyridoxamine-containing water. Studies using platelets and neutrophils from sickle cell disease mice and patients suggested that treatment with pyridoxamine reduced the activation state of platelets and neutrophils. These results suggest that pyridoxamine may be a novel therapeutic and a supplement to hydroxyurea to prevent and treat vaco-occlusion events in sickle cell disease.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Si-Yeon Jeong
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Bei Xiong
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Alan Tseng
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | | | | | - Victor R. Gordeuk
- Section of Hematology/Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Comprehensive Sickle Cell Center, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| |
Collapse
|
45
|
Cao X, Zhu Q, Xia X, Yao B, Liang S, Chen Z, Wu M. The correlation between novel peripheral blood cell ratios and 90-day mortality in patients with acute ischemic stroke. PLoS One 2020; 15:e0238312. [PMID: 32857820 PMCID: PMC7454963 DOI: 10.1371/journal.pone.0238312] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND We aimed to investigate the correlation between the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), platelet-to-neutrophil ratio (PNR), platelet-to-white blood cell ratio (PWR) and 90-day mortality in patients with acute ischemic stroke (AIS). METHODS We retrospectively included 633 patients with AIS from January 2017 to May 2018. The correlation between each indicator and the degree of neurologic deficit was assessed. Kaplan-Meier survival curves based on blood cell ratios were used to analyze the 90-day survival rate of patients with AIS. RESULTS A total of 663 patients with AIS were enrolled, of which 24 (3.6%) experienced recurrence and 13 (2.0%) died. NLR>3.23 (odds ratio; OR = 2.236; 95% confidence interval [CI], 1.472-3.397; P<0.001), PNR<31.14 (OR = 0.471; 95% CI, 0.297-0.749; P = 0.001), and PWR<20.62 (OR = 0.498; 95% CI, 0.309-0.800; P = 0.004) were associated with an unfavorable 90-day prognosis. NLR>3.23, PWR<20.62, and PNR<31.14 were associated with an increased risk of 90-day mortality. CONCLUSION PNR, PWR, and NLR were associated with the 90-day mortality of patients with AIS. Patients with high NLRs or low PWRs and PNRs may have a greater risk of mortality than other patients. These clinical indicators may help clinicians judge unfavorable prognosis early and implement the appropriate interventions.
Collapse
Affiliation(s)
- Xiaofeng Cao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qing Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Xin Xia
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Beibei Yao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Seng Liang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Zhaoyao Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Minghua Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
46
|
Wang L, Cheng Q, Peng M, Lv D, Zi W, Xu G, Liu X. The relationship between the platelet to leukocyte ratio and mechanical thrombectomy outcomes in acute ischemic stroke patients. Neurol Res 2020; 42:890-896. [PMID: 32643590 DOI: 10.1080/01616412.2020.1790868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND PURPOSE The predictive effect of blood cell ratio on ischemic event has been widely confirmed. Whether PWR and PNR can assess the risk of endovascular treatment (EVT) is largely unclear. This study aimed to investigate the prognostic value of PNR and PWR in acute ischemic stroke patients treated with EVT. METHODS Poor functional outcome was defined as Modified Rankin Scale (mRS) of 3-6 at 3 months, Symptomatic intracranial hemorrhage (sICH) was diagnosed based on CT scan and classified according to the criterial of Heidelberg Bleeding Classification. Binary logistical regression was used to analyze the relationship of PWR, PNR with functional outcome and symptomatic intracranial hemorrhage (sICH). RESULTS Patients with good prognosis had higher PNR and PWR value (29 vs. 24, P=0.002) (22 vs. 19, P=0.009), a lower rate of sICH (2.9% vs. 24.9%, P<0.001). In model 1, the lower PNR significantly associated with poor functional outcome (OR, 0.48; 95% CI 0.26-0.88; P=0.018), and sICH (OR, 0.42; 95% CI 0.19-0.91; P=0.028). The lower PWR only significantly associated with poor prognosis (OR, 0.97; 95% CI 0.94-1.00; P=0.038), and had a trend relation with sICH (OR, 0.98; 95% CI 0.94-1.02; P=0.328). In model 2 lower PNR still significantly associated with poor functional outcome (OR, 0.53; 95% CI 0.29-0.99; P=0.047), but showed a trend for predicting sICH (OR, 0.56; 95% CI 0.25-1.25; P=0.158). CONCLUSION Platelet to leukocyte ratio may be use to assess the risk of functional outcome and sICH in patients with acute anterior circulation occlusion stroke undergoing endovascular treatment in real world China.
Collapse
Affiliation(s)
- Ling Wang
- Department of Neurology, The Fourth Affiliated Hospital of Anhui Medical University , Hefei, Anhui, China.,Department of Neurology, Jinling Hospital, Medical School of Nanjing University , Nanjing, Jiangsu, China
| | - Qiantao Cheng
- Department of Neurology, The Fourth Affiliated Hospital of Anhui Medical University , Hefei, Anhui, China
| | - Min Peng
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University , Nanjing, Jiangsu, China
| | - Daping Lv
- Department of Neurology, The Fourth Affiliated Hospital of Anhui Medical University , Hefei, Anhui, China
| | - Wenjie Zi
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University , Nanjing, Jiangsu, China.,Xinqiao Hospital and the Second Affiliated Hospital, Army Medical University (Third Military Medical University) of China , Chongqing, China
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University , Nanjing, Jiangsu, China
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University , Nanjing, Jiangsu, China
| |
Collapse
|
47
|
Abstract
The term "nanotechnology" was coined by Norio Taniguchi in the 1970s to describe the manipulation of materials at the nano (10-9) scale, and the term "nanomedicine" was put forward by Eric Drexler and Robert Freitas Jr. in the 1990s to signify the application of nanotechnology in medicine. Nanomedicine encompasses a variety of systems including nanoparticles, nanofibers, surface nano-patterning, nanoporous matrices, and nanoscale coatings. Of these, nanoparticle-based applications in drug formulations and delivery have emerged as the most utilized nanomedicine system. This review aims to present a comprehensive assessment of nanomedicine approaches in vascular diseases, emphasizing particle designs, therapeutic effects, and current state-of-the-art. The expected advantages of utilizing nanoparticles for drug delivery stem from the particle's ability to (1) protect the drug from plasma-induced deactivation; (2) optimize drug pharmacokinetics and biodistribution; (3) enhance drug delivery to the disease site via passive and active mechanisms; (4) modulate drug release mechanisms via diffusion, degradation, and other unique stimuli-triggered processes; and (5) biodegrade or get eliminated safely from the body. Several nanoparticle systems encapsulating a variety of payloads have shown these advantages in vascular drug delivery applications in preclinical evaluation. At the same time, new challenges have emerged regarding discrepancy between expected and actual fate of nanoparticles in vivo, manufacturing barriers of complex nanoparticle designs, and issues of toxicity and immune response, which have limited successful clinical translation of vascular nanomedicine systems. In this context, this review will discuss challenges and opportunities to advance the field of vascular nanomedicine.
Collapse
Affiliation(s)
- Michael Sun
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
48
|
Pulavendran S, Rudd JM, Maram P, Thomas PG, Akhilesh R, Malayer JR, Chow VTK, Teluguakula N. Combination Therapy Targeting Platelet Activation and Virus Replication Protects Mice against Lethal Influenza Pneumonia. Am J Respir Cell Mol Biol 2020; 61:689-701. [PMID: 31070937 DOI: 10.1165/rcmb.2018-0196oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Excessive neutrophils recruited during influenza pneumonia contribute to severe lung pathology through induction of neutrophil extracellular traps (NETs) and release of extracellular histones. We have recently shown that activation of platelets during influenza enhances pulmonary microvascular thrombosis, leading to vascular injury and hemorrhage. Emerging evidence indicates that activated platelets also interact with neutrophils, forming neutrophil-platelet aggregates (NPAs) that contribute to tissue injury. Here, we examined neutrophil-platelet interactions and evaluated the formation of NPAs during influenza pneumonia. We also evaluated the efficacy of clopidogrel (CLP), an antagonist of the ADP-P2Y12 platelet receptor, alone or in combination with an antiviral agent (oseltamivir) against influenza infection in mice. Our studies demonstrated increased platelet activation and induction of NPAs in influenza-infected lungs, and that these NPAs led to NET release both in vitro and in vivo. Furthermore, neutrophil integrin Mac-1 (macrophage-1 antigen)-mediated platelet binding was critical for NPA formation and NET release. Administration of CLP reduced platelet activation and NPA formation but did not protect the mice against lethal influenza challenge. However, administration of CLP together with oseltamivir improved survival rates in mice compared with oseltamivir alone. The combination treatment reduced lung pathology, neutrophil influx, NPAs, NET release, and inflammatory cytokine release in infected lungs. Taken together, these results provide the first evidence that NPAs formed during influenza contribute to acute lung injury. Targeting both platelet activation and virus replication could represent an effective therapeutic option for severe influenza pneumonia.
Collapse
Affiliation(s)
- Sivasami Pulavendran
- Center for Veterinary Health Sciences and.,Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| | - Jennifer M Rudd
- Center for Veterinary Health Sciences and.,Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| | - Prasanthi Maram
- Center for Veterinary Health Sciences and.,Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee; and
| | | | | | - Vincent T K Chow
- Department of Microbiology and Immunology, School of Medicine, National University Health System, National University of Singapore, Kent Ridge, Singapore
| | - Narasaraju Teluguakula
- Center for Veterinary Health Sciences and.,Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| |
Collapse
|
49
|
Protein disulfide isomerase in cardiovascular disease. Exp Mol Med 2020; 52:390-399. [PMID: 32203104 PMCID: PMC7156431 DOI: 10.1038/s12276-020-0401-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 01/07/2023] Open
Abstract
Protein disulfide isomerase (PDI) participates in the pathogenesis of numerous diseases. Increasing evidence indicates that intravascular cell-derived PDI plays an important role in the initiation and progression of cardiovascular diseases, including thrombosis and vascular inflammation. Recent studies with PDI conditional knockout mice have advanced our understanding of the function of cell-specific PDI in disease processes. Furthermore, the identification and development of novel small-molecule PDI inhibitors has led into a new era of PDI research that transitioned from the bench to bedside. In this review, we will discuss recent findings on the regulatory role of PDI in cardiovascular disease. Efforts to untangle the functions of a large family of enzymes could lead researchers to new therapies for diverse cardiovascular diseases. Members of the protein disulfide isomerase (PDI) family chemically modify other proteins in ways that can alter both their structure and biological activity. Jaehyung Cho of the University of Illinois at Chicago, USA and coworkers have reviewed numerous studies linking PDI with cardiovascular diseases, including thrombosis, heart attack, vascular inflammation, and stroke. The authors also report progress in developing small-molecule PDI inhibitors that could yield the treatment for these conditions.
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Neutrophils priming has been long studied in vitro. Recent studies describe it in vivo. In pathophysiological conditions, complex, heterogeneous characteristics of priming are described in the last few years. RECENT FINDINGS Priming can occur systemically when insults such as sepsis or trauma result in an array of circulating mediators and circulating primed neutrophils seem to exert detrimental effects either directly, or indirectly by interacting with other cells, thereby contributing to the development of organ dysfunction. Local priming of neutrophils augments their ability to clear infection, but may also lead to local bystander tissue injury, for example, in the inflamed joint. The complexity, heterogeneity and dynamic nature of inflammatory responses and the accessibility of cells from local sites make neutrophil priming challenging to study in human disease; however, recent advances have made significant progress to this field. SUMMARY Herein, we summarize the literature regarding neutrophil priming in selected conditions. In some diseases and in the setting of specific genetic influences, the priming repertoire seems to be restricted, with only some neutrophil functions upregulated. A greater understanding of the nature of neutrophil priming and its role in human disease is required before this process becomes tractable to therapeutic intervention.
Collapse
|