1
|
Zhang Z, Zhou X, Zhou X, Cheng Z, Hu Y. Role of Platelets and Their Interaction with Immune Cells in Venous Thromboembolism. Semin Thromb Hemost 2024. [PMID: 39214148 DOI: 10.1055/s-0044-1789022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Venous thromboembolism (VTE) represents a significant global health challenge, ranking as the third leading cause of cardiovascular-related mortality. VTE pervades diverse clinical specialties, posing substantial risks to patient well-being and imposing considerable economic strains on health care systems. While platelets have long been recognized as pivotal players in hemostasis, emerging evidence underscores their multifaceted immune functions and their capacity to engage in crosstalk with other immune cells, such as neutrophils, thereby fostering immune-related thrombosis. Notably, investigations have elucidated the pivotal role of platelets in the pathogenesis of VTE. This review provides a comprehensive overview of platelet physiology, encompassing their activation, secretion dynamics, and implications in VTE. Moreover, it delineates the impact of platelet interactions with various immune cells on the initiation and progression of VTE, explores the correlation between platelet-related laboratory markers and VTE, and elucidates the role of platelets in thrombosis regression.
Collapse
Affiliation(s)
- Zhao Zhang
- Department of Hematology, Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Wuhan, China
| | - Xianghui Zhou
- Department of Hematology, Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Wuhan, China
| | - Xin Zhou
- Department of Hematology, Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Wuhan, China
| | - Zhipeng Cheng
- Department of Hematology, Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Wuhan, China
| | - Yu Hu
- Department of Hematology, Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Wuhan, China
| |
Collapse
|
2
|
Wang J, Wang X, Peng H, Dong Z, Liangpunsakul S, Zuo L, Wang H. Platelets in Alcohol-Associated Liver Disease: Interaction With Neutrophils. Cell Mol Gastroenterol Hepatol 2024; 18:41-52. [PMID: 38461963 PMCID: PMC11127035 DOI: 10.1016/j.jcmgh.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/12/2024]
Abstract
Alcohol-associated liver disease (ALD) is a major contributor to liver-related mortality globally. An increasing body of evidence underscores the pivotal role of platelets throughout the spectrum of liver injury and recovery, offering unique insights into liver homeostasis and pathobiology. Alcoholic-associated steatohepatitis is characterized by the infiltration of hepatic neutrophils. Recent studies have highlighted the extensive distance neutrophils travel through sinusoids to reach the liver injury site, relying on a platelet-paved endothelium for efficient crawling. The adherence of platelets to neutrophils is crucial for accurate migration from circulation to the inflammatory site. A gradual decline in platelet levels leads to diminished neutrophil recruitment. Platelets exhibit the ability to activate neutrophils. Platelet activation is heightened upon the release of platelet granule contents, which synergistically activate neutrophils through their respective receptors. The sequence culminates in the formation of platelet-neutrophil complexes and the release of neutrophil extracellular traps intensifies liver damage, fosters inflammatory immune responses, and triggers hepatotoxic processes. Neutrophil infiltration is a hallmark of alcohol-associated steatohepatitis, and the roles of neutrophils in ALD pathogenesis have been studied extensively, however, the involvement of platelets in ALD has received little attention. The current review consolidates recent findings on the intricate and diverse roles of platelets and neutrophils in liver pathophysiology and in ALD. Potential therapeutic strategies are highlighted, focusing on targeting platelet-neutrophil interactions and activation in ALD. The anticipation is that innovative methods for manipulating platelet and neutrophil functions will open promising avenues for future ALD therapy.
Collapse
Affiliation(s)
- Juan Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| | - Xianda Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| | - Haodong Peng
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; The First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Zijian Dong
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Li Zuo
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei, Anhui, China.
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
3
|
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
|
4
|
Platelet-Neutrophil Crosstalk in Thrombosis. Int J Mol Sci 2023; 24:ijms24021266. [PMID: 36674781 PMCID: PMC9861587 DOI: 10.3390/ijms24021266] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Platelets are essential for the formation of a haemostatic plug to prevent bleeding, while neutrophils are the guardians of our immune defences against invading pathogens. The interplay between platelets and innate immunity, and subsequent triggering of the activation of coagulation is part of the host system to prevent systemic spread of pathogen in the blood stream. Aberrant immunothrombosis and excessive inflammation can however, contribute to the thrombotic burden observed in many cardiovascular diseases. In this review, we highlight how platelets and neutrophils interact with each other and how their crosstalk is central to both arterial and venous thrombosis and in COVID-19. While targeting platelets and coagulation enables efficient antithrombotic treatments, they are often accompanied with a bleeding risk. We also discuss how novel approaches to reduce platelet-mediated recruitment of neutrophils could represent promising therapies to treat thrombosis without affecting haemostasis.
Collapse
|
5
|
Corken A, Ware J, Dai J, Arthur JM, Smyth S, Davis CL, Liu J, Harville TO, Phadnis MA, Mehta JL, Rahmatallah Y, Jain N. Platelet-Dependent Inflammatory Dysregulation in Patients with Stages 4 or 5 Chronic Kidney Disease: A Mechanistic Clinical Study. KIDNEY360 2022; 3:2036-2047. [PMID: 36591354 PMCID: PMC9802560 DOI: 10.34067/kid.0005532022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
Abstract
Background Chronic kidney disease (CKD) is characterized by dysregulated inflammation that worsens with CKD severity. The role of platelets in modulating inflammation in stage 4 or 5 CKD remains unexplored. We investigated whether there are changes in platelet-derived thromboinflammatory markers in CKD with dual antiplatelet therapy (DAPT; aspirin 81 mg/d plus P2Y12 inhibitor). Methods In a mechanistic clinical trial, we compared platelet activation markers (aggregation and surface receptor expression), circulating platelet-leukocyte aggregates, leukocyte composition (monocyte subtypes and CD11b surface expression), and plasma cytokine profile (45 analytes) of non-CKD controls (n=26) and CKD outpatients (n=48) with a glomerular filtration rate (GFR) <30 ml/min per 1.73 m2 on 2 weeks of DAPT. Results Patients with CKD demonstrated a reduced mean platelet count, elevated mean platelet volume, reduced platelet-leukocyte aggregates, reduced platelet-bound monocytes, higher total non-classic monocytes in the circulation, and higher levels of IL-1RA, VEGF, and fractalkine (all P<0.05). There were no differences in platelet activation markers between CKD and controls. Although DAPT reduced platelet aggregation in both groups, it had multifaceted effects on thromboinflammatory markers in CKD, including a reduction in PDGF levels in all CKD individuals, reductions in IL-1β and TNF-α levels in select CKD individuals, and no change in a number of other cytokines. Significant positive correlations existed for baseline IL-1β, PDGF, and TNF-α levels with older age, and for baseline TNF-α levels with presence of diabetes mellitus and worse albuminuria. Mean change in IL-1β and PDGF levels on DAPT positively correlated with younger age, mean change in TNF-α levels with higher GFR, and mean changes in PDGF, and TRAIL levels correlated with worse albuminuria. Minimum spanning trees plot of cytokines showed platelet-derived CD40L had a large reduction in weight factor after DAPT in CKD. Additionally, platelet-derived IL-1β and PDGF were tightly correlated with other cytokines, with IL-1β as the hub cytokine. Conclusions Attenuated interactions between platelets and leukocytes in the CKD state coincided with no change in platelet activation status, an altered differentiation state of monocytes, and heightened inflammatory markers. Platelet-derived cytokines were one of the central cytokines in patients with CKD that were tightly correlated with others. DAPT had multifaceted effects on thromboinflammation, suggesting that there is platelet-dependent and -independent inflammation in stage 4 or 5 CKD.
Collapse
Affiliation(s)
- Adam Corken
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jerry Ware
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Junqiang Dai
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - John M. Arthur
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas,Central Arkansas Veterans Health Care System, Little Rock, Arkansas
| | - Susan Smyth
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Clayton L. Davis
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Juan Liu
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Terry O. Harville
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas,Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Milind A. Phadnis
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Jawahar L. Mehta
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas,Central Arkansas Veterans Health Care System, Little Rock, Arkansas
| | - Yasir Rahmatallah
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Nishank Jain
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas,Central Arkansas Veterans Health Care System, Little Rock, Arkansas
| |
Collapse
|
6
|
Altered platelet functions during treatment with apremilast for psoriatic arthritis: A case report. Curr Res Transl Med 2022; 70:103358. [PMID: 35724504 DOI: 10.1016/j.retram.2022.103358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/05/2022] [Indexed: 01/31/2023]
|
7
|
de Pins B, Mendes T, Giralt A, Girault JA. The Non-receptor Tyrosine Kinase Pyk2 in Brain Function and Neurological and Psychiatric Diseases. Front Synaptic Neurosci 2021; 13:749001. [PMID: 34690733 PMCID: PMC8527176 DOI: 10.3389/fnsyn.2021.749001] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/14/2021] [Indexed: 12/28/2022] Open
Abstract
Pyk2 is a non-receptor tyrosine kinase highly enriched in forebrain neurons. Pyk2 is closely related to focal adhesion kinase (FAK), which plays an important role in sensing cell contacts with extracellular matrix and other extracellular signals controlling adhesion and survival. Pyk2 shares some of FAK’s characteristics including recruitment of Src-family kinases after autophosphorylation, scaffolding by interacting with multiple partners, and activation of downstream signaling pathways. Pyk2, however, has the unique property to respond to increases in intracellular free Ca2+, which triggers its autophosphorylation following stimulation of various receptors including glutamate NMDA receptors. Pyk2 is dephosphorylated by the striatal-enriched phosphatase (STEP) that is highly expressed in the same neuronal populations. Pyk2 localization in neurons is dynamic, and altered following stimulation, with post-synaptic and nuclear enrichment. As a signaling protein Pyk2 is involved in multiple pathways resulting in sometimes opposing functions depending on experimental models. Thus Pyk2 has a dual role on neurites and dendritic spines. With Src family kinases Pyk2 participates in postsynaptic regulations including of NMDA receptors and is necessary for specific types of synaptic plasticity and spatial memory tasks. The diverse functions of Pyk2 are also illustrated by its role in pathology. Pyk2 is activated following epileptic seizures or ischemia-reperfusion and may contribute to the consequences of these insults whereas Pyk2 deficit may contribute to the hippocampal phenotype of Huntington’s disease. Pyk2 gene, PTK2B, is associated with the risk for late-onset Alzheimer’s disease. Studies of underlying mechanisms indicate a complex contribution with involvement in amyloid toxicity and tauopathy, combined with possible functional deficits in neurons and contribution in microglia. A role of Pyk2 has also been proposed in stress-induced depression and cocaine addiction. Pyk2 is also important for the mobility of astrocytes and glioblastoma cells. The implication of Pyk2 in various pathological conditions supports its potential interest for therapeutic interventions. This is possible through molecules inhibiting its activity or increasing it through inhibition of STEP or other means, depending on a precise evaluation of the balance between positive and negative consequences of Pyk2 actions.
Collapse
Affiliation(s)
- Benoit de Pins
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Tiago Mendes
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Jean-Antoine Girault
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| |
Collapse
|
8
|
Okabayashi K, Kanai S, Katakura F, Takeuchi R, Yamauchi T, Nakayama S, Kinoshita R, Koie H, Narita T. Activation of canine neutrophils by platelet-activating factor. Vet Immunol Immunopathol 2021; 241:110336. [PMID: 34649042 DOI: 10.1016/j.vetimm.2021.110336] [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: 05/07/2021] [Revised: 09/14/2021] [Accepted: 10/02/2021] [Indexed: 10/20/2022]
Abstract
Neutrophils are essential for innate immunity as the first line of defence. Neutrophils act as phagocytic white blood cells to kill bacteria and other microorganisms. A strong respiratory burst of neutrophils, dependent on reactive oxygen species, is produced during phagocytosis. Platelet-activating factor (PAF) is a signalling molecule with several prominent roles in tissue injury, inflammation, and platelet aggregation. However, the detailed mechanisms and intracellular signalling pathways involved in PAF-mediated neutrophil activation remain unclear. Here, we investigated the effect of PAF on changes in calcium concentration ([Ca2+]i) and oxygen radical (O2-) generation in activating canine neutrophils. We further evaluated these effects of PAF with inhibition of G protein-coupled receptors using the specific inhibitor suramin. Blood samples were collected from a total of five dogs and neutrophils were isolated. PAF stimulation of canine neutrophils caused an increase in [Ca2+]i as well as the generation of O2-, and the PAF receptor was sensitive to suramin. The results suggested that PAF stimulation of canine neutrophils may cause Ca2+ influx from the endoplasmic reticulum into the cytoplasm (as the first wave) and then trigger store-operated Ca2+ entry (as the second wave), which is an important intracellular signal transduction pathway for neutrophil activation. Furthermore, O2- generation by PAF stimulation may depend on the intracellular signalling pathway, with increasing inositol trisphosphate levels and [Ca2+]i via G protein-coupled receptors. The finding that PAF-activating platelet aggregation is involved in canine neutrophil activation suggests a close relationship between haemostasis and neutrophil activation in dogs, offering new insight into the response to infection.
Collapse
Affiliation(s)
- Ken Okabayashi
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa, 252-0880, Japan
| | - Shuichiro Kanai
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa, 252-0880, Japan
| | - Fumihiko Katakura
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa, 252-0880, Japan
| | - Riku Takeuchi
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa, 252-0880, Japan
| | - Takashi Yamauchi
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa, 252-0880, Japan
| | - Shunya Nakayama
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa, 252-0880, Japan
| | - Rie Kinoshita
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa, 252-0880, Japan
| | - Hiroshi Koie
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa, 252-0880, Japan
| | - Takanori Narita
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa, 252-0880, Japan.
| |
Collapse
|
9
|
Totani L, Amore C, Piccoli A, Dell'Elba G, Di Santo A, Plebani R, Pecce R, Martelli N, Rossi A, Ranucci S, De Fino I, Moretti P, Bragonzi A, Romano M, Evangelista V. Type-4 Phosphodiesterase (PDE4) Blockade Reduces NETosis in Cystic Fibrosis. Front Pharmacol 2021; 12:702677. [PMID: 34566635 PMCID: PMC8456009 DOI: 10.3389/fphar.2021.702677] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/26/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophilic inflammation is a key determinant of cystic fibrosis (CF) lung disease. Neutrophil-derived free DNA, released in the form of extracellular traps (NETs), significantly correlates with impaired lung function in patients with CF, underlying their pathogenetic role in CF lung disease. Thus, specific approaches to control NETosis of neutrophils migrated into the lungs may be clinically relevant in CF. We investigated the efficacy of phosphodiesterase (PDE) type-4 inhibitors, in vitro, on NET release by neutrophils from healthy volunteers and individuals with CF, and in vivo, on NET accumulation and lung inflammation in mice infected with Pseudomonas aeruginosa. PDE4 blockade curbed endotoxin-induced NET production and preserved cellular integrity and apoptosis in neutrophils, from healthy subjects and patients with CF, challenged with endotoxin, in vitro. The pharmacological effects of PDE4 inhibitors were significantly more evident on CF neutrophils. In a mouse model of Pseudomonas aeruginosa chronic infection, aerosol treatment with roflumilast, a selective PDE4 inhibitor, gave a significant reduction in free DNA in the BALF. This was accompanied by reduced citrullination of histone H3 in neutrophils migrated into the airways. Roflumilast-treated mice showed a significant improvement in weight recovery. Our study provides the first evidence that PDE4 blockade controls NETosis in vitro and in vivo, in CF-relevant models. Since selective PDE4 inhibitors have been recently approved for the treatment of COPD and psoriasis, our present results encourage clinical trials to test the efficacy of this class of drugs in CF.
Collapse
Affiliation(s)
- Licia Totani
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro (CH), Mozzagrogna, Italy
| | - Concetta Amore
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro (CH), Mozzagrogna, Italy
| | - Antonio Piccoli
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro (CH), Mozzagrogna, Italy
| | - Giuseppe Dell'Elba
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro (CH), Mozzagrogna, Italy
| | - Angelo Di Santo
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro (CH), Mozzagrogna, Italy
| | - Roberto Plebani
- Laboratory of Molecular Medicine, Centre for Advanced Studies and Technology (CAST), Department of Medical Oral and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Romina Pecce
- Laboratory of Molecular Medicine, Centre for Advanced Studies and Technology (CAST), Department of Medical Oral and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Nicola Martelli
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro (CH), Mozzagrogna, Italy
| | - Alice Rossi
- Infection and Cystic Fibrosis Unit, Division of Immunology Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serena Ranucci
- Infection and Cystic Fibrosis Unit, Division of Immunology Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ida De Fino
- Infection and Cystic Fibrosis Unit, Division of Immunology Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Moretti
- Cystic Fibrosis Centre, S. Liberatore Hospital, Atri, Italy
| | - Alessandra Bragonzi
- Infection and Cystic Fibrosis Unit, Division of Immunology Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mario Romano
- Laboratory of Molecular Medicine, Centre for Advanced Studies and Technology (CAST), Department of Medical Oral and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Virgilio Evangelista
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro (CH), Mozzagrogna, Italy
| |
Collapse
|
10
|
Kao TI, Chen PJ, Wang YH, Tseng HH, Chang SH, Wu TS, Yang SH, Lee YT, Hwang TL. Bletinib ameliorates neutrophilic inflammation and lung injury by inhibiting Src family kinase phosphorylation and activity. Br J Pharmacol 2021; 178:4069-4084. [PMID: 34131920 PMCID: PMC8518616 DOI: 10.1111/bph.15597] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/07/2021] [Accepted: 04/29/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Neutrophil overactivation is crucial in the pathogenesis of acute lung injury (ALI). Bletinib (3,3'-dihydroxy-2',6'-bis(p-hydroxybenzyl)-5-methoxybibenzyl), a natural bibenzyl, extracted from the Bletilla plant, exhibits anti-inflammatory, antibacterial, and antimitotic effects. In this study, we evaluated the therapeutic effects of bletinib in human neutrophilic inflammation and LPS-mediated ALI in mice. EXPERIMENTAL APPROACH In human neutrophils activated with the formyl peptide (fMLP), we assessed integrin expression, superoxide anion production, degranulation, neutrophil extracellular trap (NET) formation, and adhesion through flow cytometry, spectrophotometry, and immunofluorescence microscopy. Immunoblotting was used to measure phosphorylation of Src family kinases (SFKs) and downstream proteins. Finally, a LPS-induced ALI model in male BALB/c mice was used to investigate the potential therapeutic effects of bletinib treatment. KEY RESULTS In activated human neutrophils, bletinib reduced degranulation, respiratory burst, NET formation, adhesion, migration, and integrin expression; suppressed the enzymic activity of SFKs, including Src, Lyn, Fgr, and Hck; and inhibited the phosphorylation of SFKs as well as Vav and Bruton's tyrosine kinase (Btk). In mice with ALI, the pulmonary sections demonstrated considerable amelioration of prominent inflammatory changes, such as haemorrhage, pulmonary oedema, and neutrophil infiltration, after bletinib treatment. CONCLUSION AND IMPLICATIONS Bletinib regulates neutrophilic inflammation by inhibiting the SFK-Btk-Vav pathway. Bletinib ameliorates LPS-induced ALI in mice. Further biochemical optimisation of bletinib may be a promising strategy for the development of novel therapeutic agents for inflammatory diseases.
Collapse
Affiliation(s)
- Ting-I Kao
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Chinese Internal Medicine, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Jen Chen
- Department of Cosmetic Science, Providence University, Taichung, Taiwan
| | - Yi-Hsuan Wang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Hui Tseng
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Hsin Chang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Tian-Shung Wu
- Department of Chemistry, National Cheng Kung University, Tainan, Taiwan
| | - Sien-Hung Yang
- Division of Chinese Internal Medicine, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Yen-Tung Lee
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Cosmetic Science, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Chinese Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
11
|
Chen WA, Fletcher HM, Payne KJ, Aka S, Thornburg MB, Gheorghe JD, Safi SB, Shavlik D, Oyoyo U, Boskovic DS. Platelet and neutrophil responses to Porphyromonas gingivalis in human whole blood. Mol Oral Microbiol 2021; 36:202-213. [PMID: 33811483 DOI: 10.1111/omi.12336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/01/2021] [Accepted: 03/30/2021] [Indexed: 01/19/2023]
Abstract
Porphyromonas gingivalis is a causative agent for periodontal disease. Binding of platelets to this gram-negative anaerobe can regulate host hemostatic (thrombus forming) and immune (neutrophil interacting) responses during bacterial infection. Additionally, in response to bacterial pathogens neutrophils can release their DNA, forming highly prothrombotic neutrophil extracellular traps (NETs), which then further enhance platelet responses. This study evaluates the role of P. gingivalis on platelet expression of CD62P, platelet-neutrophil interactions, and labeled neutrophil-associated DNA. Human whole blood was preincubated with varying P. gingivalis concentrations, with or without subsequent addition of adenosine diphosphate (ADP). Flow cytometry was employed to measure platelet expression of CD62P using PerCP-anti-CD61 and PE-anti-CD62P, platelet-neutrophil interactions using PerCP-anti-CD61 and FITC-anti-CD16, and the release of neutrophil DNA using FITC-anti-CD16 and Sytox Blue labeling. Preincubation with a high (6.25 × 106 CFU/mL) level of P. gingivalis significantly increased platelet expression of CD62P in ADP treated and untreated whole blood. In addition, platelet-neutrophil interactions were significantly increased after ADP stimulation, following 5-22 min preincubation of blood with high P. gingivalis CFU. However, in the absence of added ADP, platelet-neutrophil interactions increased in a manner dependent on the preincubation time with P. gingivalis. Moreover, after ADP addition, 16 min preincubation of whole blood with P. gingivalis led to increased labeling of neutrophil-associated DNA. Taken together, the results suggest that the presence of P. gingivalis alters platelet and neutrophil responses to increase platelet activation, platelet interactions with neutrophils, and the level of neutrophil antimicrobial NETs.
Collapse
Affiliation(s)
- William A Chen
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Hansel M Fletcher
- Division of Microbiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Kimberly J Payne
- Division of Anatomy, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Sheryl Aka
- Department of Pathology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Melanie B Thornburg
- Department of Pathology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Joseph D Gheorghe
- Department of Pathology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Shahnaj Binte Safi
- Department of Epidemiology and Biostatistics, School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - David Shavlik
- Department of Epidemiology and Biostatistics, School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - Udochukwu Oyoyo
- Department of Dental Education Services, School of Dentistry, Loma Linda University, Loma Linda, CA, USA
| | - Danilo S Boskovic
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
12
|
Cai C, Sun H, Hu L, Fan Z. Visualization of integrin molecules by fluorescence imaging and techniques. ACTA ACUST UNITED AC 2021; 45:229-257. [PMID: 34219865 PMCID: PMC8249084 DOI: 10.32604/biocell.2021.014338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Integrin molecules are transmembrane αβ heterodimers involved in cell adhesion, trafficking, and signaling. Upon activation, integrins undergo dynamic conformational changes that regulate their affinity to ligands. The physiological functions and activation mechanisms of integrins have been heavily discussed in previous studies and reviews, but the fluorescence imaging techniques -which are powerful tools for biological studies- have not. Here we review the fluorescence labeling methods, imaging techniques, as well as Förster resonance energy transfer assays used to study integrin expression, localization, activation, and functions.
Collapse
Affiliation(s)
- Chen Cai
- Department of Immunology, School of Medicine, UConn Health, Farmington, 06030, USA
| | - Hao Sun
- Department of Medicine, University of California, San Diego, La Jolla, 92093, USA
| | - Liang Hu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450051, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, 06030, USA
| |
Collapse
|
13
|
Godino C, Scotti A, Maugeri N, Mancini N, Fominskiy E, Margonato A, Landoni G. Antithrombotic therapy in patients with COVID-19? -Rationale and Evidence. Int J Cardiol 2021; 324:261-266. [PMID: 33002521 PMCID: PMC7521414 DOI: 10.1016/j.ijcard.2020.09.064] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/09/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022]
Abstract
In patients with severe or critical Coronavirus disease 2019 (COVID-19) manifestations, a thromboinflammatory syndrome, with diffuse microvascular thrombosis, is increasingly evident as the final step of pro-inflammatory cytokines storm. Actually, no proven effective therapies for novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection exist. Preliminary observations on anticoagulant therapy appear to be associated with better outcomes in moderate and severe COVID-19 patients with signs of coagulopathy and in those requiring mechanical ventilation. The pathophysiology underlying the prothrombotic state elicited by SARS-CoV-2 outlines possible protective mechanisms of antithrombotic therapy (in primis anticoagulants) for this viral illness. The indications for antiplatelet/anticoagulant use (prevention, prophylaxis, therapy) are guided by the clinical context and the COVID-19 severity. We provide a practical approach on antithrombotic therapy management for COVID-19 patients from a multidisciplinary point of view.
Collapse
Affiliation(s)
- Cosmo Godino
- Clinical Cardiology Unit, Faculty of Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Andrea Scotti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua Medical School, Padua, Italy
| | - Norma Maugeri
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicasio Mancini
- Laboratory of Medical Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy; Faculty of Medicine, Vita Salute San Raffaele University, Milan, Italy
| | - Evgeny Fominskiy
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Margonato
- Clinical Cardiology Unit, Faculty of Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy; Faculty of Medicine, Vita Salute San Raffaele University, Milan, Italy
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy; Faculty of Medicine, Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
14
|
Dehghani T, Panitch A. Endothelial cells, neutrophils and platelets: getting to the bottom of an inflammatory triangle. Open Biol 2020; 10:200161. [PMID: 33050789 PMCID: PMC7653352 DOI: 10.1098/rsob.200161] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
Severe fibrotic and thrombotic events permeate the healthcare system, causing suffering for millions of patients with inflammatory disorders. As late-state consequences of chronic inflammation, fibrosis and thrombosis are the culmination of pathological interactions of activated endothelium, neutrophils and platelets after vessel injury. Coupling of these three cell types ensures a pro-coagulant, cytokine-rich environment that promotes the capture, activation and proliferation of circulating immune cells and recruitment of key pro-fibrotic cell types such as myofibroblasts. As the first responders to sterile inflammatory injury, it is important to understand how endothelial cells, neutrophils and platelets help create this environment. There has been a growing interest in this intersection over the past decade that has helped shape the development of therapeutics to target these processes. Here, we review recent insights into how neutrophils, platelets and endothelial cells guide the development of pathological vessel repair that can also result in underlying tissue fibrosis. We further discuss recent efforts that have been made to translate this knowledge into therapeutics and provide perspective as to how a compound or combination therapeutics may be most efficacious when tackling fibrosis and thrombosis that is brought upon by chronic inflammation.
Collapse
Affiliation(s)
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Drive, GBSF 2303, Davis, CA, USA
| |
Collapse
|
15
|
Qi X, Fan M, Huang N, Zhang X, Liu J, Li X, Sun R. Saikosaponin d contributed to cancer chemotherapy induced neutropenia therapy by promoting neutrophil differentiation via activation CBL-dependent ERK pathway. Pharmacol Res 2020; 160:105149. [PMID: 32822868 DOI: 10.1016/j.phrs.2020.105149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 12/16/2022]
Abstract
Cancer chemotherapy induced neutropenia (CCIN) is one of the most common toxicity caused by cytotoxic anticancer agents. Despite granulocyte colony-stimulating factor (GCSF) is widely used in clinical practice, the infection and infection-related mortality rate is still high for lack of functionally mature neutrophils. Saikosaponin d (SSD) is one of the major bioactive constituents of Radix Bupleuri (RB), which exerts immune-modulatory properties. We explored the function of SSD in CCIN therapy, we found that SSD contributed to generate functional mature neutrophils which capable of fighting infection both in vitro and in vivo. Network pharmacology was employed to explore the mechanism, 61 signal pathways might play an important role in CCIN treatment. Western Blot was employed to further confirm the potential pathway involved. We found CBL-ERK1/2 pathway was activated by SSD, followed by upregulating PU.1 and CEBPβ expression and leading to neutrophil differentiation. Our findings suggest a natural regimen SSD which could regenerate microbicidal neutrophils to effectively reduce CCIN-associated infection via activating CBL-ERK1/2, providing a rationale for future therapeutic approaches.
Collapse
Affiliation(s)
- Xiaotian Qi
- Institute of Advanced Medical Research, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Mengyue Fan
- Institute of Advanced Medical Research, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, China
| | - Nana Huang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, China
| | - Xinyu Zhang
- Institute of Advanced Medical Research, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, China
| | - Jing Liu
- Institute of Advanced Medical Research, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, China
| | - Xiaoyu Li
- Department of Medical Pathomorphology, Shandong Academy of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Rong Sun
- Institute of Advanced Medical Research, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, China.
| |
Collapse
|
16
|
Canino J, Guidetti GF, Galgano L, Vismara M, Minetti G, Torti M, Canobbio I. The proline-rich tyrosine kinase Pyk2 modulates integrin-mediated neutrophil adhesion and reactive oxygen species generation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118799. [PMID: 32693110 DOI: 10.1016/j.bbamcr.2020.118799] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/03/2020] [Accepted: 07/14/2020] [Indexed: 01/05/2023]
Abstract
Neutrophils are first responders in infection and inflammation. They are able to roll, adhere and transmigrate through the endothelium to reach the site of infection, where they fight pathogens through secretion of granule contents, production of reactive oxygen species, extrusion of neutrophil extracellular traps, and phagocytosis. In this study we explored the role of the non-receptor focal adhesion kinase Pyk2 in neutrophil adhesion and activation. Using a specific Pyk2 pharmacological inhibitor, PF-4594755, as well as Pyk2-deficient murine neutrophils, we found that Pyk2 is activated upon integrin αMβ2-mediated neutrophil adhesion to fibrinogen. This process is triggered by Src family kinases-mediated phosphorylation and supported by Pyk2 autophosphorylation on Y402. In neutrophil adherent to fibrinogen, Pyk2 activates PI3K-dependent pathways promoting the phosphorylation of Akt and of its downstream effector GSK3. Pyk2 also dynamically regulates MAP kinases in fibrinogen-adherent neutrophils, as it stimulates p38MAPK but negatively regulates ERK1/2. Pharmacological inhibition of Pyk2 significantly prevented adhesion of human neutrophils to fibrinogen, and neutrophils from Pyk2-knockout mice showed a reduced ability to adhere compared to wildtype cells. Accordingly, neutrophil adhesion to fibrinogen was reduced upon inhibition of p38MAPK but potentiated by ERK1/2 inhibition. Neutrophil adherent to fibrinogen, but not to polylysine, were able to produce ROS upon lipopolysaccharide challenge and ROS production was completely suppressed upon inhibition of Pyk2. By contrast PMA-induced ROS production by neutrophil adherent to either fibrinogen or polylysine was independent from Pyk2. Altogether these results demonstrate that Pyk2 is an important effector in the coordinated puzzle regulating neutrophil adhesion and activation.
Collapse
Affiliation(s)
- Jessica Canino
- Department of Biology and Biotechnology, University of Pavia, Italy; Scuola Universitaria Superiore, IUSS, Pavia, Italy
| | | | - Luca Galgano
- Department of Biology and Biotechnology, University of Pavia, Italy; Scuola Universitaria Superiore, IUSS, Pavia, Italy
| | - Mauro Vismara
- Department of Biology and Biotechnology, University of Pavia, Italy
| | | | - Mauro Torti
- Department of Biology and Biotechnology, University of Pavia, Italy
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, University of Pavia, Italy.
| |
Collapse
|
17
|
Nguyen GT, Shaban L, Mack M, Swanson KD, Bunnell SC, Sykes DB, Mecsas J. SKAP2 is required for defense against K. pneumoniae infection and neutrophil respiratory burst. eLife 2020; 9:56656. [PMID: 32352382 PMCID: PMC7250567 DOI: 10.7554/elife.56656] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/29/2020] [Indexed: 12/11/2022] Open
Abstract
Klebsiella pneumoniae is a respiratory, blood, liver, and bladder pathogen of significant clinical concern. We show that the adaptor protein, SKAP2, is required for protection against K. pneumoniae (ATCC 43816) pulmonary infections. Skap2-/- mice had 100-fold higher bacterial burden when compared to wild-type and burden was controlled by SKAP2 expression in innate immune cells. Skap2-/- neutrophils and monocytes were present in infected lungs, and the neutrophils degranulated normally in response to K. pneumoniae infection in mice; however, K. pneumoniae-stimulated reactive oxygen species (ROS) production in vitro was abolished. K. pneumoniae-induced neutrophil ROS response required the activity of SFKs, Syk, Btk, PLCγ2, and PKC. The loss of SKAP2 significantly hindered the K. pneumoniae-induced phosphorylation of SFKs, Syk, and Pyk2 implicating SKAP2 as proximal to their activation in pathogen-signaling pathways. In conclusion, SKAP2-dependent signaling in neutrophils is essential for K. pneumoniae-activated ROS production and for promoting bacterial clearance during infection. Klebsiella pneumoniae is a type of bacteria that can cause life-threatening infections – including pneumonia, blood stream infections, and urinary tract infections – in hospitalized patients. These infections can be difficult to treat because some K. pneumoniae are resistant to antibiotics. The bacteria are normally found in the human intestine, and they do not usually cause infections in healthy people. This implies that healthy people’s immune systems are better able to fend off K. pneumoniae infections; learning how could help scientists develop new ways to treat or prevent infections in hospitalized patients. In healthy people, a type of immune cell called neutrophils are the first line of defense against bacterial infections. Several different proteins are needed to activate neutrophils, including a protein called SKAP2. But the role of this protein in fighting K. pneumoniae infections is not clear. To find out what role SKAP2 plays in the defense against pneumonia caused by K. pneumoniae, Nguyen et al. compared infections in mice with and without the protein. Mice lacking SKAP2 in their white blood cells had more bacteria in their lungs than normal mice. The experiments showed that neutrophils from mice with SKAP2 produce a burst of chemicals called “reactive oxygen species”, which can kill bacteria. But neutrophils without the protein do not. Without SKAP2, several proteins that help produce reactive oxygen species do not work. Understanding the role of SKAP2 in fighting infections may help scientists better understand the immune system. This could help clinicians to treat conditions that cause it to be hyperactive or ineffective. More studies are needed to determine if SKAP2 works the same way in human neutrophils and if it works against all types of K. pneumoniae. If it does, then scientists might be able use this information to develop therapies that help the immune system fight infections.
Collapse
Affiliation(s)
- Giang T Nguyen
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, United States
| | - Lamyaa Shaban
- Graduate Program in Molecular Microbiology, Tufts Graduate School of Biomedical Sciences, Boston, United States
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Kenneth D Swanson
- Brain Tumor Center and Neuro-Oncology Unit, Department of Neurology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, United States
| | - Stephen C Bunnell
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, United States.,Department of Immunology, School of Medicine, Tufts University, Boston, United States
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, United States
| | - Joan Mecsas
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, United States.,Graduate Program in Molecular Microbiology, Tufts Graduate School of Biomedical Sciences, Boston, United States.,Department of Molecular Biology and Microbiology, School of Medicine, Tufts University, Boston, United States
| |
Collapse
|
18
|
Chaudhary PK, Han JS, Jee Y, Lee SH, Kim S. Pyk2 downstream of G 12/13 pathways regulates platelet shape change through RhoA/p160 ROCK. Biochem Biophys Res Commun 2020; 526:738-743. [PMID: 32265034 DOI: 10.1016/j.bbrc.2020.03.130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/16/2020] [Accepted: 03/20/2020] [Indexed: 11/29/2022]
Abstract
Rho/Rho-kinase downstream of G12/13 plays an important role in the regulation of calcium-independent platelet shape change. We have previously shown that proline-rich tyrosine kinase 2 (Pyk2) is activated downstream of G12/13 pathways. In this study, we evaluated the role of Pyk2 in G12/13-induced platelet shape change. We used low concentrations of YFLLRNP, a heptapeptide binding to protease-activated receptor 1 (PAR1), or PAR4-activating peptide AYPGKF in the presence of Gαq inhibitor YM254890 to selectively stimulate G12/13 pathways. We found that G12/13-induced platelet shape change was completely inhibited in the presence of Pyk2 inhibitors AG17 and TAT-Pyk2-CT, suggesting an important role of Pyk2 in platelet shape change. In addition, AYPGKF-induced shape change in Gq -/- platelets was completely inhibited in the presence of AG17 or RhoA/p160ROCK inhibitor Y27632, confirming the role of Pyk2 in RhoA-dependent shape change. Furthermore, AYPGKF-induced platelet aggregation and dense granule secretion were inhibited by blocking Pyk2 or RhoA. Finally, G12/13-induced myosin phosphatase target subunit 1 (MYPT1) phosphorylation was inhibited by AG17, confirming that Pyk2 regulates RhoA/p160ROCK activation in platelets. These results demonstrate that Pyk2 downstream of G12/13 pathways regulates platelet shape change as well as platelet aggregation and dense granule secretion through the regulation of RhoA/p160ROCK.
Collapse
Affiliation(s)
| | - Jeung-Sul Han
- College of Agriculture & Life Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Youngheun Jee
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, South Korea
| | - Seung-Hun Lee
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea
| | - Soochong Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.
| |
Collapse
|
19
|
McDonald B, Dunbar M. Platelets and Intravascular Immunity: Guardians of the Vascular Space During Bloodstream Infections and Sepsis. Front Immunol 2019; 10:2400. [PMID: 31681291 PMCID: PMC6797619 DOI: 10.3389/fimmu.2019.02400] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022] Open
Abstract
Despite their humble origins as anuclear fragments of megakaryocytes, platelets have emerged as versatile mediators of thrombosis and immunity. The diverse spectrum of platelet functions are on full display during the host response to severe infection and sepsis, with platelets taking center-stage in the intravascular immune response to blood-borne pathogens. Platelets are endowed with a comprehensive armamentarium of pathogen detection systems that enable them to function as sentinels in the bloodstream for rapid identification of microbial invasion. Through both autonomous anti-microbial effector functions and collaborations with other innate immune cells, platelets orchestrate a complex intravascular immune defense system that protects against bacterial dissemination. As with any powerful immune defense system, dysregulation of platelet-mediated intravascular immunity can lead to profound collateral damage to host cells and tissues, resulting in sepsis-associated organ dysfunction. In this article, the cellular and molecular contributions of platelets to intravascular immune defenses in sepsis will be reviewed, including the roles of platelets in surveillance of the microcirculation and elicitation of protective anti-bacterial responses. Mechanisms of platelet-mediated thromboinflammatory organ dysfunction will be explored, with linkages to clinical biomarkers of platelet homeostasis that aid in the diagnosis and prognostication of human sepsis. Lastly, we discuss novel therapeutic opportunities that take advantage of our evolving understanding of platelets and intravascular immunity in severe infection.
Collapse
Affiliation(s)
- Braedon McDonald
- Department of Critical Care Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mary Dunbar
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
20
|
Guidetti GF, Torti M, Canobbio I. Focal Adhesion Kinases in Platelet Function and Thrombosis. Arterioscler Thromb Vasc Biol 2019; 39:857-868. [DOI: 10.1161/atvbaha.118.311787] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The focal adhesion kinase family includes 2 homolog members, FAK and Pyk2 (proline-rich tyrosine kinase 2), primarily known for their roles in nucleated cells as regulators of cytoskeletal dynamics and cell adhesion. FAK and Pyk2 are also expressed in megakaryocytes and platelets and are activated by soluble agonists and on adhesion to the subendothelial matrix. Despite high sequence homology and similar molecular organization, FAK and Pyk2 play different roles in platelet function. Whereas FAK serves mostly as a traditional focal adhesion kinase activated downstream of integrins, Pyk2 coordinates multiple signals from different receptors. FAK, but not Pyk2, is involved in megakaryocyte maturation and platelet production. In circulating platelets, FAK is recruited by integrin αIIbβ3 to regulate hemostasis, whereas it plays minimal roles in thrombosis. By contrast, Pyk2 is implicated in platelet activation and is an important regulator of thrombosis. The direct activation of Pyk2 by calcium ions provides a connection between GPCRs (G-protein coupled receptors) and Src family kinases. In this review, we provide the comprehensive overview of >20 years of investigations on the role and regulation of focal adhesion kinases in blood platelets, highlighting common and distinctive features of FAK and Pyk2 in hemostasis and thrombosis.
Collapse
Affiliation(s)
| | - Mauro Torti
- From the Department of Biology and Biotechnology, University of Pavia, Italy
| | - Ilaria Canobbio
- From the Department of Biology and Biotechnology, University of Pavia, Italy
| |
Collapse
|
21
|
|
22
|
Borsig L. Selectins in cancer immunity. Glycobiology 2018; 28:648-655. [PMID: 29272415 DOI: 10.1093/glycob/cwx105] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022] Open
Abstract
Selectins are vascular adhesion molecules that mediate physiological responses such as inflammation, immunity and hemostasis. During cancer progression, selectins promote various steps enabling the interactions between tumor cells and the blood constituents, including platelets, endothelial cells and leukocytes. Selectins are carbohydrate-binding molecules that bind to sialylated, fucosylated glycan structures. The increased selectin ligand expression on tumor cells correlates with enhanced metastasis and poor prognosis for cancer patients. While, many studies focused on the role of selectin as a mediator of tumor cell adhesion and extravasation during metastasis, there is evidence for selectins to activate signaling cascade that regulates immune responses within a tumor microenvironment. L-Selectin binding induces activation of leukocytes, which can be further modulated by selectin-mediated interactions with platelets and endothelial cells. Selectin ligand on leukocytes, PSGL-1, triggers intracellular signaling in leukocytes that are induced through platelet's P-selectin or endothelial E-selectin binding. In this review, I summarize the evidence for selectin-induced immune modulation in cancer progression that represents a possible target for controlling tumor immunity.
Collapse
Affiliation(s)
- Lubor Borsig
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Winterthurerstrasse 190, Zurich, Switzerland
| |
Collapse
|
23
|
Finsterbusch M, Norman MU, Hall P, Kitching AR, Hickey MJ. Platelet retention in inflamed glomeruli occurs via selective prolongation of interactions with immune cells. Kidney Int 2018; 95:363-374. [PMID: 30522769 DOI: 10.1016/j.kint.2018.08.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 08/13/2018] [Accepted: 08/30/2018] [Indexed: 01/16/2023]
Abstract
Platelet-leukocyte interactions promote acute glomerulonephritis. However, neither the nature of the interactions between platelets and immune cells nor the capacity of platelets to promote leukocyte activation has been characterized in this condition. We used confocal intravital microscopy to define the interactions of platelets with neutrophils, monocytes, and endothelial cells in glomerular capillaries in mice. In the absence of inflammation, platelets underwent rapid on/off interactions with immune cells. During glomerulonephritis induced by in situ immune complex formation, platelets that interacted with neutrophils or monocytes, but not with other intraglomerular cells, were retained in the glomerulus for prolonged durations. Depletion of platelets inhibited both neutrophil recruitment and activation. Inhibition of platelet activating factor reduced neutrophil recruitment without impacting reactive oxygen species generation, while blocking CXC chemokine ligand 7 (CXCL7) reduced both responses. In contrast, inhibition of the adenosine diphosphate and thromboxane A2 pathways inhibited neutrophil reactive oxygen species generation without affecting neutrophil adhesion. Thus, platelet retention in glomerular capillaries following immune complex deposition stems from prolongation of platelet interactions with immune cells but not other substrates. Pro-inflammatory mediators play divergent roles in promoting neutrophil retention and activation in glomerular capillaries.
Collapse
Affiliation(s)
- Michaela Finsterbusch
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - M Ursula Norman
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Pam Hall
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Medical Centre, Clayton, Victoria, Australia; Department of Paediatric Nephrology, Monash Medical Centre, Clayton, Victoria, Australia
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia.
| |
Collapse
|
24
|
Rossaint J, Margraf A, Zarbock A. Role of Platelets in Leukocyte Recruitment and Resolution of Inflammation. Front Immunol 2018; 9:2712. [PMID: 30515177 PMCID: PMC6255980 DOI: 10.3389/fimmu.2018.02712] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/02/2018] [Indexed: 12/30/2022] Open
Abstract
Platelets are most often recognized for their crucial role in the control of acute hemorrhage. However, current research has greatly expanded the appreciation of platelets beyond their contribution to primary hemostasis, indicating that platelets also actively participate in leukocyte recruitment and the regulation of the host defense in response to exogenous pathogens and sterile injury. Early recruitment of leukocytes, especially neutrophils, is the evolutionary stronghold of the innate immune response to successfully control exogenous infections. Platelets have been shown to physically interact with different leukocyte subsets during inflammatory processes. This interaction holds far-reaching implications for the leukocyte recruitment into peripheral tissues as well as the regulation of leukocyte cell autonomous functions, including the formation and liberation of neutrophil extracellular traps. These functions critically depend on the interaction of platelets with leukocytes. The host immune response and leukocyte recruitment must be tightly regulated to avoid excessive tissue and organ damage and to avoid chronification of inflammation. Thus, platelet-leukocyte interactions and the resulting leukocyte activation and recruitment also underlies tight regulation by several inherited feedback mechanisms to limit the extend of vascular inflammation and to protect the host from collateral damage caused by overshooting immune system activation. After the acute inflammatory phase has been overcome the host defense response must eventually be terminated to allow for resolution from inflammation and restoration of tissue and organ function. Besides their essential role for leukocyte recruitment and the initiation and propagation of vascular inflammation, platelets have lately also been implicated in the resolution process. Here, their contribution to phagocyte clearance, T cell recruitment and macrophage reprogramming is also of outmost importance. This review will focus on the role of platelets in leukocyte recruitment during the initiation of the host defense and we will also discuss the participation of platelets in the resolution process after acute inflammation.
Collapse
Affiliation(s)
- Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Andreas Margraf
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany.,Interdisciplinary Centre for Clinical Research, University Hospital Münster, Münster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
25
|
Nakayama H, Nagafuku M, Suzuki A, Iwabuchi K, Inokuchi JI. The regulatory roles of glycosphingolipid-enriched lipid rafts in immune systems. FEBS Lett 2018; 592:3921-3942. [PMID: 30320884 DOI: 10.1002/1873-3468.13275] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 01/04/2023]
Abstract
Lipid rafts formed by glycosphingolipids (GSLs) on cellular membranes play important roles in innate and adaptive immunity. Lactosylceramide (LacCer) forms lipid rafts on plasma and granular membranes of human neutrophils. These LacCer-enriched lipid rafts bind directly to pathogenic components, such as pathogenic fungi-derived β-glucan and Mycobacteria-derived lipoarabinomannan via carbohydrate-carbohydrate interactions, and mediate innate immune responses to these pathogens. In contrast, a-series and o-series gangliosides form distinct rafts on CD4+ and CD8+ T cell subsets, respectively, contributing to the respective functions of these cells and stimulating adaptive immune responses through T cell receptors. These findings suggest that gangliosides play indispensable roles in T cell selection and activation. This Review introduces the involvement of GSL-enriched lipid rafts in innate and adaptive immunity.
Collapse
Affiliation(s)
- Hitoshi Nakayama
- Laboratory of Biochemistry, Juntendo University Faculty of Health Care and Nursing, Urayasu, Japan.,Institute for Environmental and Gender-specific Medicine, Juntendo University Graduate School of Medicine, Urayasu, Japan
| | - Masakazu Nagafuku
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Akemi Suzuki
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kazuhisa Iwabuchi
- Laboratory of Biochemistry, Juntendo University Faculty of Health Care and Nursing, Urayasu, Japan.,Institute for Environmental and Gender-specific Medicine, Juntendo University Graduate School of Medicine, Urayasu, Japan.,Infection Control Nursing, Juntendo University Graduate School of Health Care and Nursing, Urayasu, Japan
| | - Jin-Ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
26
|
Deppermann C, Kubes P. Start a fire, kill the bug: The role of platelets in inflammation and infection. Innate Immun 2018; 24:335-348. [PMID: 30049243 PMCID: PMC6830908 DOI: 10.1177/1753425918789255] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/16/2018] [Accepted: 06/26/2018] [Indexed: 11/19/2022] Open
Abstract
Platelets are the main players in thrombosis and hemostasis; however they also play important roles during inflammation and infection. Through their surface receptors, platelets can directly interact with pathogens and immune cells. Platelets form complexes with neutrophils to modulate their capacities to produce reactive oxygen species or form neutrophil extracellular traps. Furthermore, they release microbicidal factors and cytokines that kill pathogens and influence the immune response, respectively. Platelets also maintain the vascular integrity during inflammation by a mechanism that is different from classical platelet activation. In this review we summarize the current knowledge about how platelets interact with the innate immune system during inflammation and infection and highlight recent advances in the field.
Collapse
Affiliation(s)
- Carsten Deppermann
- Calvin, Phoebe and Joan Snyder Institute for Chronic
Diseases, University of Calgary, Calgary, AB, Canada
| | - Paul Kubes
- Calvin, Phoebe and Joan Snyder Institute for Chronic
Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
27
|
Manfredi AA, Ramirez GA, Rovere-Querini P, Maugeri N. The Neutrophil's Choice: Phagocytose vs Make Neutrophil Extracellular Traps. Front Immunol 2018. [PMID: 29515586 PMCID: PMC5826238 DOI: 10.3389/fimmu.2018.00288] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neutrophils recognize particulate substrates of microbial or endogenous origin and react by sequestering the cargo via phagocytosis or by releasing neutrophil extracellular traps (NETs) outside the cell, thus modifying and alerting the environment and bystander leukocytes. The signals that determine the choice between phagocytosis and the generation of NETs are still poorly characterized. Neutrophils that had phagocytosed bulky particulate substrates, such as apoptotic cells and activated platelets, appear to be “poised” in an unresponsive state. Environmental conditions, the metabolic, adhesive and activation state of the phagocyte, and the size of and signals associated with the tethered phagocytic cargo influence the choice of the neutrophils, prompting either phagocytic clearance or the generation of NETs. The choice is dichotomic and apparently irreversible. Defects in phagocytosis may foster the intravascular generation of NETs, thus promoting vascular inflammation and morbidities associated with diseases characterized by defective phagocytic clearance, such as systemic lupus erythematosus. There is a strong potential for novel treatments based on new knowledge of the events determining the inflammatory and pro-thrombotic function of inflammatory leukocytes.
Collapse
Affiliation(s)
- Angelo A Manfredi
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Giuseppe A Ramirez
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Patrizia Rovere-Querini
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Norma Maugeri
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| |
Collapse
|
28
|
Iwabuchi K. Gangliosides in the Immune System: Role of Glycosphingolipids and Glycosphingolipid-Enriched Lipid Rafts in Immunological Functions. Methods Mol Biol 2018; 1804:83-95. [PMID: 29926405 DOI: 10.1007/978-1-4939-8552-4_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Although individuals are constantly exposed to infectious agents, these agents are generally resisted by the innate and acquired immune systems. Both the innate and acquired immune systems protect against invading organisms, but they differ functionally in several ways. The innate immune system is the body's inborn defense mechanism and the first line of defense against invading organisms, such as bacteria, fungi, and viruses. Glycosphingolipids (GSLs), which are expressed on the outer leaflet of plasma membranes (Murate et al., J Cell Sci 128(8):1627-1638, 2015), are involved in both innate and acquired immunity (Inokuchi et al., Biochim Biophys Acta 1851(1):98-106, 2015; Nakayama et al., Arch Immunol Ther Exp (Warsz) 61(3):217-228, 2013; Rueda, Br J Nutr 98(Suppl 1):S68-73, 2007; Popa and Portoukalian, Pathol Biol (Paris) 51(5):253-255, 2003).Recent studies have indicated that innate immunity is not a "nonspecific" immune system. Large numbers of viruses, bacteria, and bacterial toxins have been reported to bind to host surface carbohydrates, a number of which are components of GSLs (Schengrund, Biochem Pharmacol 65(5):699-707, 2003). Binding studies have also demonstrated that some glycolipids function as receptors for microorganisms and bacterial toxins (Yates and Rampersaud, Ann N Y Acad Sci 845:57-71, 1998). These findings clearly indicate that GSLs are involved in host-pathogen interactions.GSLs are composed of hydrophobic ceramide and hydrophilic sugar moieties (Hakomori, Annu Rev Biochem 50:733-764, 1980). The ceramide moiety of sphingolipids and the cholesterol sterol-ring system are thought to interact via hydrogen bonds and hydrophobic van der Waal's forces (Mukherjee and Maxfield, Annu Rev Cell Dev Biol 20:839-866, 2004). Additional hydrophilic cis interactions among GSL headgroups have been found to promote their lateral associations with surrounding lipid and protein membrane components. These interactions result in the separation in cell membranes of lipid rafts, which are lipid domains rich in GSLs, cholesterol, glycosylphosphatidylinositol (GPI)-anchored proteins and membrane-anchored signaling molecules (Pike, J Lipid Res 47(7):1597-1598, 2006). These GSL-enriched lipid rafts play important roles in immunological functions (Inokuchi et al., Biochim Biophys Acta 1851(1):98-106, 2015; Iwabuchi et al., Mediators Inflamm 2015:120748, 2015; Anderson and Roche, Biochim Biophys Acta 1853(4):775-780, 2015; Zuidscherwoude et al., J Leukoc Biol 95(2):251-263, 2014; Dykstra et al., Annu Rev Immunol 21:457-481, 2003). This introductory chapter describes the roles of GSLs and their lipid rafts in the immune system.
Collapse
Affiliation(s)
- Kazuhisa Iwabuchi
- Infection Control Nursing, Graduate School of Health Care and Nursing, Juntendo University, Chiba, Japan.
- Institute for Environmental and Gender Specific Medicine, Graduate school of Medicine, Juntendo University, Chiba, Japan.
| |
Collapse
|
29
|
Nurden A. Platelets, inflammation and tissue regeneration. Thromb Haemost 2017; 105 Suppl 1:S13-33. [DOI: 10.1160/ths10-11-0720] [Citation(s) in RCA: 469] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 02/04/2011] [Indexed: 12/20/2022]
Abstract
SummaryBlood platelets have long been recognised to bring about primary haemostasis with deficiencies in platelet production and function manifesting in bleeding while upregulated function favourises arterial thrombosis. Yet increasing evidence indicates that platelets fulfil a much wider role in health and disease. First, they store and release a wide range of biologically active substances including the panoply of growth factors, chemokines and cytokines released from α-granules. Membrane budding gives rise to microparticles (MPs), another active participant within the blood stream. Platelets are essential for the innate immune response and combat infection (viruses, bacteria, micro-organisms). They help maintain and modulate inflammation and are a major source of pro-inflammatory molecules (e.g. P-selectin, tissue factor, CD40L, metalloproteinases). As well as promoting coagulation, they are active in fibrinolysis; wound healing, angiogenesis and bone formation as well as in maternal tissue and foetal vascular remodelling. Activated platelets and MPs intervene in the propagation of major diseases. They are major players in atherosclerosis and related diseases, pathologies of the central nervous system (Alzheimers disease, multiple sclerosis), cancer and tumour growth. They participate in other tissue-related acquired pathologies such as skin diseases and allergy, rheumatoid arthritis, liver disease; while, paradoxically, autologous platelet-rich plasma and platelet releasate are being used as an aid to promote tissue repair and cellular growth. The above mentioned roles of platelets are now discussed.
Collapse
|
30
|
Hosseini E, Ghasemzadeh M. Intravascular leukocyte migration through platelet thrombi: directing leukocytes to sites of vascular injury. Thromb Haemost 2017; 113:1224-35. [DOI: 10.1160/th14-08-0662] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 01/13/2015] [Indexed: 12/15/2022]
Abstract
SummaryLeukocytes recruitment to thrombi supports an intimate cellular interaction leading to the enhancement of pro-coagulant functions and pro-inflammatory responses at site of vascular injury. Recent observations of neutrophil extracellular traps (NETs) formation and its mutual reactions with platelet thrombi adds more clinical interest to the growing body of knowledge in the field of platelet-leukocyte crosstalk. However, having considered thrombus as a barrier between leukocytes and injured endothelium, the full inflammatory roles of these cells during thrombosis is still ill defined. The most recent observation of neutrophils migration into the thrombi is a phenomenon that highlights the inflammatory functions of leukocytes at the site of injury. It has been hypothesised that leukocytes migration might be associated with the conveyance of highly reactive pro-inflammatory and/or procoagulant mediators to sites of vascular injury. In addition, the evidence of neutrophils migration into arterial thrombi following traumatic and ischaemia-reperfusion injury highlights the already described role of these cells in atherosclerosis. Regardless of the mechanisms behind leukocyte migration, whether these migrated cells benefit normal homeostasis by their involvement in wound healing and vascular rebuilding or they increase unwilling inflammatory responses, could be of interest for future researches that provide new insight into biological importance of leukocyte recruitment to thrombi.
Collapse
|
31
|
Singhal R, Chawla S, Rathore DK, Bhasym A, Annarapu GK, Sharma V, Seth T, Guchhait P. Development of pro-inflammatory phenotype in monocytes after engulfing Hb-activated platelets in hemolytic disorders. Clin Immunol 2016; 175:133-142. [PMID: 28039017 DOI: 10.1016/j.clim.2016.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/31/2016] [Accepted: 12/03/2016] [Indexed: 01/31/2023]
Abstract
Monocytes and macrophage combat infections and maintain homeostatic balance by engulfing microbes and apoptotic cells, and releasing inflammatory cytokines. Studies have described that these cells develop anti-inflammatory properties upon recycling the free-hemoglobin (Hb) in hemolytic conditions. While investigating the phenotype of monocytes in two hemolytic disorders-paroxysmal nocturnal hemoglobinuria (PNH) and sickle cell disease (SCD), we observed a high number of pro-inflammatory (CD14+CD16hi) monocytes in these patients. We further investigated in vitro the phenotype of these monocytes and found an estimated 55% of CD14+ cells were transformed into the CD14+CD16hi subset after engulfing Hb-activated platelets. The CD14+CD16hi monocytes, which were positive for both intracellular Hb and CD42b (platelet marker), secreted significant amounts of TNF-α and IL-1β, unlike monocytes treated with only free Hb, which secreted more IL-10. We have shown recently the presence of a high number of Hb-bound hyperactive platelets in patients with both diseases, and further investigated if the monocytes engulfed these activated platelets in vivo. As expected, we found 95% of CD14+CD16hi monocytes with both intracellular Hb and CD42b in both diseases, and they expressed high TNF-α. Furthermore our data showed that these monocytes whether from patients or developed in vitro after treatment with Hb-activated platelets, secreted significant amounts of tissue factor. Besides, these CD14+CD16hi monocytes displayed significantly decreased phagocytosis of E. coli. Our study therefore suggests that this alteration of monocyte phenotype may play a role in the increased propensity to pro-inflammatory/coagulant complications observed in these hemolytic disorders-PNH and SCD.
Collapse
Affiliation(s)
- Rashi Singhal
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India; Department of Biotechnology, Manipal University, Karnataka, India
| | - Sheetal Chawla
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Deepak K Rathore
- Translational Health Science & Technology Institute, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Angika Bhasym
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India; Department of Biotechnology, Manipal University, Karnataka, India
| | - Gowtham K Annarapu
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India; Department of Biotechnology, Manipal University, Karnataka, India
| | - Vandana Sharma
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, India
| | - Tulika Seth
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
32
|
Thrombocytopenia is associated with a dysregulated host response in critically ill sepsis patients. Blood 2016; 127:3062-72. [PMID: 26956172 DOI: 10.1182/blood-2015-11-680744] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/12/2016] [Indexed: 12/15/2022] Open
Abstract
Preclinical studies have suggested that platelets influence the host response during sepsis. We sought to assess the association of admission thrombocytopenia with the presentation, outcome, and host response in patients with sepsis. Nine hundred thirty-one consecutive sepsis patients were stratified according to platelet counts (very low <50 × 10(9)/L, intermediate-low 50 × 10(9) to 99 × 10(9)/L, low 100 × 10(9) to 149 × 10(9)/L, or normal 150 × 10(9) to 399 × 10(9)/L) on admission to the intensive care unit. Sepsis patients with platelet counts <50 × 10(9)/L and 50 × 10(9) to 99 × 10(9)/L presented with higher Acute Physiology and Chronic Health Evaluation scores and more shock. Both levels of thrombocytopenia were independently associated with increased 30-day mortality (hazard ratios with 95% confidence intervals 2.00 [1.32-3.05] and 1.72 [1.22-2.44], respectively). To account for baseline differences besides platelet counts, propensity matching was performed, after which the association between thrombocytopenia and the host response was tested, as evaluated by measuring 17 plasma biomarkers indicative of activation and/or dysregulation of pathways implicated in sepsis pathogenesis and by whole genome blood leukocyte expression profiling. In the propensity matched cohort, platelet counts < 50 × 10(9)/L were associated with increased cytokine levels and enhanced endothelial cell activation. All thrombocytopenic groups showed evidence of impaired vascular integrity, whereas coagulation activation was similar between groups. Blood microarray analysis revealed a distinct gene expression pattern in sepsis patients with <50 × 10(9)/L platelets, showing reduced signaling in leukocyte adhesion and diapedesis and increased complement signaling. These data show that admission thrombocytopenia is associated with enhanced mortality and a more disturbed host response during sepsis independent of disease severity, thereby providing clinical validity to animal studies on the role of platelets in severe infection.
Collapse
|
33
|
Andrade SS, Gouvea IE, Silva MCC, Castro ED, de Paula CAA, Okamoto D, Oliveira L, Peres GB, Ottaiano T, Facina G, Nazário ACP, Campos AHJFM, Paredes-Gamero EJ, Juliano M, da Silva IDCG, Oliva MLV, Girão MJBC. Cathepsin K induces platelet dysfunction and affects cell signaling in breast cancer - molecularly distinct behavior of cathepsin K in breast cancer. BMC Cancer 2016; 16:173. [PMID: 26931461 PMCID: PMC4774035 DOI: 10.1186/s12885-016-2203-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/17/2016] [Indexed: 11/12/2022] Open
Abstract
Background Breast cancer comprises clinically and molecularly distinct tumor subgroups that differ in cell histology and biology and show divergent clinical phenotypes that impede phase III trials, such as those utilizing cathepsin K inhibitors. Here we correlate the epithelial-mesenchymal-like transition breast cancer cells and cathepsin K secretion with activation and aggregation of platelets. Cathepsin K is up-regulated in cancer cells that proteolyze extracellular matrix and contributes to invasiveness. Although proteolytically activated receptors (PARs) are activated by proteases, the direct interaction of cysteine cathepsins with PARs is poorly understood. In human platelets, PAR-1 and −4 are highly expressed, but PAR-3 shows low expression and unclear functions. Methods Platelet aggregation was monitored by measuring changes in turbidity. Platelets were immunoblotted with anti-phospho and total p38, Src-Tyr-416, FAK-Tyr-397, and TGFβ monoclonal antibody. Activation was measured in a flow cytometer and calcium mobilization in a confocal microscope. Mammary epithelial cells were prepared from the primary breast cancer samples of 15 women with Luminal-B subtype to produce primary cells. Results We demonstrate that platelets are aggregated by cathepsin K in a dose-dependent manner, but not by other cysteine cathepsins. PARs-3 and −4 were confirmed as the cathepsin K target by immunodetection and specific antagonists using a fibroblast cell line derived from PARs deficient mice. Moreover, through co-culture experiments, we show that platelets activated by cathepsin K mediated the up-regulation of SHH, PTHrP, OPN, and TGFβ in epithelial-mesenchymal-like cells from patients with Luminal B breast cancer. Conclusions Cathepsin K induces platelet dysfunction and affects signaling in breast cancer cells. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2203-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sheila Siqueira Andrade
- Departments of Gynecology of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil. .,Charitable Association of Blood Collection - COLSAN, São Paulo, SP, 04080-006, Brazil. .,Department of Gynecology, Cellular Gynecology Laboratory, Universidade Federal de São Paulo, Rua Napoleão de Barros, 608, CEP 04024-002, São Paulo, Brazil.
| | - Iuri Estrada Gouvea
- Biophysics of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | | | - Eloísa Dognani Castro
- Biochemistry of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Cláudia A A de Paula
- Biochemistry of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Debora Okamoto
- Biophysics of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Lilian Oliveira
- Biophysics of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Giovani Bravin Peres
- Biochemistry of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Tatiana Ottaiano
- Biochemistry of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Gil Facina
- Departments of Gynecology of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | | | - Antonio Hugo J F M Campos
- Department of Pathology, AC Camargo Hospital Biobank, A C Camargo Cancer Center - Antonio Prudente Foundation, São Paulo, SP, 01509-010, Brazil.
| | | | - Maria Juliano
- Biophysics of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Ismael D C G da Silva
- Departments of Gynecology of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Maria Luiza V Oliva
- Biochemistry of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil.
| | - Manoel J B C Girão
- Departments of Gynecology of Universidade Federal de São Paulo, São Paulo, SP, 04024-002, Brazil. .,Charitable Association of Blood Collection - COLSAN, São Paulo, SP, 04080-006, Brazil.
| |
Collapse
|
34
|
Totani L, Amore C, Di Santo A, Dell'Elba G, Piccoli A, Martelli N, Tenor H, Beume R, Evangelista V. Roflumilast inhibits leukocyte-platelet interactions and prevents the prothrombotic functions of polymorphonuclear leukocytes and monocytes. J Thromb Haemost 2016; 14:191-204. [PMID: 26484898 DOI: 10.1111/jth.13173] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 10/04/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED ESSENTIALS: Thrombosis is a major comorbidity in patients with chronic obstructive pulmonary disease (COPD). Roflumilast is a selective phosphodiesterase type-4 (PDE4) inhibitor approved for treatment of severe COPD. PDE4 blockade by roflumilast inhibits prothrombotic functions of neutrophils and monocytes. PDE4 inhibitors may reduce thrombotic risk in COPD as well as in other vascular diseases. BACKGROUND Roflumilast, an oral selective phosphodiesterase type 4 inhibitor, is approved for the treatment of severe chronic obstructive pulmonary disease (COPD). A recent meta-analysis of trials on COPD revealed that treatment with roflumilast was associated with a significant reduction in the rate of major cardiovascular events. The mechanisms of this effect remain unknown. OBJECTIVES We tested the hypothesis that roflumilast N-oxide (RNO), the active metabolite of roflumilast, curbs the molecular mechanisms required for leukocyte-platelet (PLT) interactions and prevents the prothrombotic functions of polymorphonuclear leukocytes (PMNs) and monocytes (MNs). METHODS Using well-characterized in vitro models, we analysed the effects of RNO on: (i) PMN adhesiveness; (ii) the release of neutrophil extracellular traps (NETs); and (iii) tissue factor expression in MNs. Key biochemical events underlying the inhibitory effects of RNO were defined. RESULTS AND CONCLUSIONS In PMNs, RNO prevented phosphoinositide 3-kinase (PI3K)-dependent phosphorylation of Akt on Ser473, and Src family kinase (SFK)-mediated Pyk2 phosphorylation on Tyr579-580, while inducing protein kinase A-mediated phosphorylation of C-terminal Src kinase, the major negative regulator of SFKs. Modulation of these signaling pathways by RNO resulted in a significant impairment of PMN adhesion to activated PLTs or human umbilical vein endothelial cells, mainly mediated by inhibition of the adhesive function of Mac-1. Moreover RNO curbed SFK/PI3K-mediated NET release by PMNs adherent on fibrinogen-coated surfaces. In MNs interacting with activated PLTs, RNO curbed PI3K-mediated expression of tissue factor. The efficacy of RNO was significantly potentiated by formoterol, a long acting β-adrenergic receptor agonist. This study reveals novel antithrombotic activities by which roflumilast may exert protective effects against cardiovascular comorbodities in COPD.
Collapse
Affiliation(s)
- L Totani
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - C Amore
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - A Di Santo
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - G Dell'Elba
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - A Piccoli
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - N Martelli
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - H Tenor
- Takeda Pharmaceuticals International GmbH, Glattpark-Opfikon, Switzerland
| | - R Beume
- Takeda Pharmaceuticals International GmbH, Glattpark-Opfikon, Switzerland
| | - V Evangelista
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| |
Collapse
|
35
|
Ampofo E, Müller I, Dahmke IN, Eichler H, Montenarh M, Menger MD, Laschke MW. Role of protein kinase CK2 in the dynamic interaction of platelets, leukocytes and endothelial cells during thrombus formation. Thromb Res 2015; 136:996-1006. [DOI: 10.1016/j.thromres.2015.08.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 07/28/2015] [Accepted: 08/29/2015] [Indexed: 10/23/2022]
|
36
|
Herrmann JM, Meyle J. Neutrophil activation and periodontal tissue injury. Periodontol 2000 2015; 69:111-27. [DOI: 10.1111/prd.12088] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
37
|
Rossaint J, Zarbock A. Platelets in leucocyte recruitment and function. Cardiovasc Res 2015; 107:386-95. [PMID: 25712962 DOI: 10.1093/cvr/cvv048] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/08/2015] [Indexed: 11/14/2022] Open
Abstract
Platelets have a longstanding recognition as an essential cellular component of the coagulation system. However, substantial research over the last decade has added another important aspect to platelet function in that they are also an integral part of the innate immune system. Complex organisms are facing a constant threat of infections by invading pathogens, and they have developed a sophisticated and elegant measure to combat this threat, namely the immune system. Leucocyte recruitment to sites of infections is an essential step at the forefront of the immune response. Platelets have been shown to be involved in several steps of this process and they are an integrated connecting element among haemostasis, host defence, and additional immunological functions (e.g. neutrophil extracellular traps formation). However, the immune system also requires a tight regulation, as an overshooting immune response carries the risk of harming the host itself. This review aims at highlighting the unique features and molecular mechanisms that allow for the interactions of platelets and leucocytes and the regulation of this process. Furthermore, this article identifies the functional relevance of these events for the immune response.
Collapse
Affiliation(s)
- Jan Rossaint
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, Münster 48149, Germany Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Alexander Zarbock
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, Münster 48149, Germany Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
38
|
Li J, Kim K, Barazia A, Tseng A, Cho J. Platelet-neutrophil interactions under thromboinflammatory conditions. Cell Mol Life Sci 2015; 72:2627-43. [PMID: 25650236 DOI: 10.1007/s00018-015-1845-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 01/07/2015] [Accepted: 01/26/2015] [Indexed: 12/11/2022]
Abstract
Platelets primarily mediate hemostasis and thrombosis, whereas leukocytes are responsible for immune responses. Since platelets interact with leukocytes at the site of vascular injury, thrombosis and vascular inflammation are closely intertwined and occur consecutively. Recent studies using real-time imaging technology demonstrated that platelet-neutrophil interactions on the activated endothelium are an important determinant of microvascular occlusion during thromboinflammatory disease in which inflammation is coupled to thrombosis. Although the major receptors and counter receptors have been identified, it remains poorly understood how heterotypic platelet-neutrophil interactions are regulated under disease conditions. This review discusses our current understanding of the regulatory mechanisms of platelet-neutrophil interactions in thromboinflammatory disease.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, University of Illinois College of Medicine, 835 S. Wolcott Ave, E403, Chicago, IL, 60612, USA
| | | | | | | | | |
Collapse
|
39
|
Stolla M, Refaai MA, Heal JM, Spinelli SL, Garraud O, Phipps RP, Blumberg N. Platelet transfusion - the new immunology of an old therapy. Front Immunol 2015; 6:28. [PMID: 25699046 PMCID: PMC4313719 DOI: 10.3389/fimmu.2015.00028] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/14/2015] [Indexed: 01/14/2023] Open
Abstract
Platelet transfusion has been a vital therapeutic approach in patients with hematologic malignancies for close to half a century. Randomized trials show that prophylactic platelet transfusions mitigate bleeding in patients with acute myeloid leukemia. However, even with prophylactic transfusions, as many as 75% of patients, experience hemorrhage. While platelet transfusion efficacy is modest, questions and concerns have arisen about the risks of platelet transfusion therapy. The acknowledged serious risks of platelet transfusion include viral transmission, bacterial sepsis, and acute lung injury. Less serious adverse effects include allergic and non-hemolytic febrile reactions. Rare hemolytic reactions have occurred due to a common policy of transfusing without regard to ABO type. In the last decade or so, new concerns have arisen; platelet-derived lipids are implicated in transfusion-related acute lung injury after transfusion. With the recognition that platelets are immune cells came the discoveries that supernatant IL-6, IL-27 sCD40L, and OX40L are closely linked to febrile reactions and sCD40L with acute lung injury. Platelet transfusions are pro-inflammatory, and may be pro-thrombotic. Anti-A and anti-B can bind to incompatible recipient or donor platelets and soluble antigens, impair hemostasis and thus increase bleeding. Finally, stored platelet supernatants contain biological mediators such as VEGF and TGF-β1 that may compromise the host versus tumor response. This is particularly of concern in patients receiving many platelet transfusions, as for acute leukemia. New evidence suggests that removing stored supernatant will improve clinical outcomes. This new view of platelets as pro-inflammatory and immunomodulatory agents suggests that innovative approaches to improving platelet storage and pre-transfusion manipulations to reduce toxicity could substantially improve the efficacy and safety of this long-employed therapy.
Collapse
Affiliation(s)
- Moritz Stolla
- Department of Pathology and Laboratory Medicine, School of Medicine and Dentistry, University of Rochester Medical Center , Rochester, NY , USA
| | - Majed A Refaai
- Department of Pathology and Laboratory Medicine, School of Medicine and Dentistry, University of Rochester Medical Center , Rochester, NY , USA
| | - Joanna M Heal
- Department of Pathology and Laboratory Medicine, School of Medicine and Dentistry, University of Rochester Medical Center , Rochester, NY , USA
| | - Sherry L Spinelli
- Department of Pathology and Laboratory Medicine, School of Medicine and Dentistry, University of Rochester Medical Center , Rochester, NY , USA
| | - Olivier Garraud
- Etablissement Francais du Sang Auvergne-Loire, Universite de Lyon , Saint-Etienne , France
| | - Richard P Phipps
- Department of Pathology and Laboratory Medicine, School of Medicine and Dentistry, University of Rochester Medical Center , Rochester, NY , USA ; Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester Medical Center , Rochester, NY , USA ; Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester Medical Center , Rochester, NY , USA ; Department of Medicine, School of Medicine and Dentistry, University of Rochester Medical Center , Rochester, NY , USA
| | - Neil Blumberg
- Department of Pathology and Laboratory Medicine, School of Medicine and Dentistry, University of Rochester Medical Center , Rochester, NY , USA
| |
Collapse
|
40
|
Carrim N, Walsh TG, Consonni A, Torti M, Berndt MC, Metharom P. Role of focal adhesion tyrosine kinases in GPVI-dependent platelet activation and reactive oxygen species formation. PLoS One 2014; 9:e113679. [PMID: 25415317 PMCID: PMC4240642 DOI: 10.1371/journal.pone.0113679] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 10/29/2014] [Indexed: 01/22/2023] Open
Abstract
Background We have previously shown the presence of a TRAF4/p47phox/Hic5/Pyk2 complex associated with the platelet collagen receptor, GPVI, consistent with a potential role of this complex in GPVI-dependent ROS formation. In other cell systems, NOX-dependent ROS formation is facilitated by Pyk2, which along with its closely related homologue FAK are known to be activated and phosphorylated downstream of ligand binding to GPVI. Aims To evaluate the relative roles of Pyk2 and FAK in GPVI-dependent ROS formation and to determine their location within the GPVI signaling pathway. Methods and Results Human and mouse washed platelets (from WT or Pyk2 KO mice) were pre-treated with pharmacological inhibitors targeting FAK or Pyk2 (PF-228 and Tyrphostin A9, respectively) and stimulated with the GPVI-specific agonist, CRP. FAK, but not Pyk2, was found to be essential for GPVI-dependent ROS production and aggregation. Subsequent human platelet studies with PF-228 confirmed FAK is essential for GPVI-mediated phosphatidylserine exposure, α-granule secretion (P-selectin (CD62P) surface expression) and integrin αIIbβ3 activation. To determine the precise location of FAK within the GPVI pathway, we analyzed the effect of PF-228 inhibition in CRP-stimulated platelets in conjunction with immunoprecipitation and pulldown analysis to show that FAK is downstream of Lyn, Spleen tyrosine kinase (Syk), PI3-K and Bruton's tyrosine kinase (Btk) and upstream of Rac1, PLCγ2, Ca2+ release, PKC, Hic-5, NOX1 and αIIbβ3 activation. Conclusion Overall, these data suggest a novel role for FAK in GPVI-dependent ROS formation and platelet activation and elucidate a proximal signaling role for FAK within the GPVI pathway.
Collapse
Affiliation(s)
- Naadiya Carrim
- Department of Experimental Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tony G. Walsh
- Department of Experimental Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alessandra Consonni
- Laboratories of Biochemistry, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Mauro Torti
- Laboratories of Biochemistry, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Michael C. Berndt
- Department of Experimental Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Pat Metharom
- Department of Experimental Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Australia
- * E-mail:
| |
Collapse
|
41
|
Totani L, Piccoli A, Dell'Elba G, Concetta A, Di Santo A, Martelli N, Federico L, Pamuklar Z, Smyth SS, Evangelista V. Phosphodiesterase type 4 blockade prevents platelet-mediated neutrophil recruitment at the site of vascular injury. Arterioscler Thromb Vasc Biol 2014; 34:1689-96. [PMID: 24925970 DOI: 10.1161/atvbaha.114.303939] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Platelet-neutrophil interactions play a key role in cardiovascular disease and inflammatory processes. Src family kinases mediate P-selectin glycoprotein ligand-1-Mac-1 cross talk necessary for firm platelet-neutrophil adhesion. Because Src family kinase activity can be regulated by cAMP-dependent pathways, in this work, we evaluated the role of phosphodiesterases in the signaling events that are required to sustain platelet-neutrophil interactions and neutrophil recruitment at the site of vascular injury. APPROACH AND RESULTS In neutrophils exposed to P-selectin, selective phosphodiesterase 4 (PDE4) inhibition prevented Src family kinase-mediated phosphorylation of the proline-rich tyrosine kinase 2 on Tyr579/Tyr580. The effects of PDE4 inhibition required protein kinase A, likely through protein kinase A-mediated activation of COOH-terminal Src kinase, a major negative regulator of Src family kinases. PDE4, but not other phosphodiesterase inhibitors, reduced platelet-neutrophil conjugates as well as neutrophil firm adhesion on spread platelets under flow conditions. The effect of PDE4 inhibition on neutrophil adhesion was primarily mediated by downregulation of P-selectin-induced activation of Mac-1. In a murine model of endovascular injury, selective inhibition of PDE4 significantly reduced neutrophil recruitment at the site of vascular damage. CONCLUSIONS This study identifies PDE4 as a central node in the signaling network that mediates platelet-neutrophil adhesion and suggests that pharmacological inhibition of PDE4 may represent a novel therapeutic avenue for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Licia Totani
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Antonio Piccoli
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Giuseppe Dell'Elba
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Amore Concetta
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Angelomaria Di Santo
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Nicola Martelli
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Lorenzo Federico
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Zehra Pamuklar
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Susan S Smyth
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Virgilio Evangelista
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.).
| |
Collapse
|
42
|
Platelets protect from septic shock by inhibiting macrophage-dependent inflammation via the cyclooxygenase 1 signalling pathway. Nat Commun 2014; 4:2657. [PMID: 24150174 DOI: 10.1038/ncomms3657] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/23/2013] [Indexed: 12/31/2022] Open
Abstract
Although it has long been known that patients with sepsis often have thrombocytopenia and that septic patients with severe thrombocytopenia have a poor prognosis and higher mortality, the role of platelets in the pathogenesis of sepsis is poorly understood. Here we report a protective role of platelets in septic shock. We show that experimental thrombocytopenia induced by intraperitoneal injection of an anti-glycoprotein Ibα monoclonal antibody increases mortality and aggravates organ failure, whereas transfusion of platelets reduces mortality in lipopolysaccharide-induced endotoxemia and a bacterial infusion mouse sepsis model. Plasma concentrations of proinflammatory cytokines TNF-α and IL-6 are elevated by thrombocytopenia and decreased by platelet transfusion in septic mice. Furthermore, we identify that platelets protect from septic shock by inhibiting macrophage-dependent inflammation via the COX1/PGE₂/EP4-dependent pathway. Thus, these findings demonstrate a previously unappreciated role for platelets in septic shock and suggest that platelet transfusion may be effective in treating severely septic patients.
Collapse
|
43
|
Flannagan RS, Canton J, Furuya W, Glogauer M, Grinstein S. The phosphatidylserine receptor TIM4 utilizes integrins as coreceptors to effect phagocytosis. Mol Biol Cell 2014; 25:1511-22. [PMID: 24623723 PMCID: PMC4004599 DOI: 10.1091/mbc.e13-04-0212] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
TIM4 is a receptor for phosphatidylserine that mediates engulfment of apoptotic cells. Remarkably, it does not require its cytosolic or transmembrane domains to mediate phagocytosis. TIM4 associates with integrins that serve as signal-transducing coreceptors. T-cell immunoglobulin mucin protein 4 (TIM4), a phosphatidylserine (PtdSer)-binding receptor, mediates the phagocytosis of apoptotic cells. How TIM4 exerts its function is unclear, and conflicting data have emerged. To define the mode of action of TIM4, we used two distinct but complementary approaches: 1) we compared bone marrow–derived macrophages from wild-type and TIM4−/− mice, and 2) we heterologously expressed TIM4 in epithelioid AD293 cells, which rendered them competent for engulfment of PtdSer-bearing targets. Using these systems, we demonstrate that rather than serving merely as a tether, as proposed earlier by others, TIM4 is an active participant in the phagocytic process. Furthermore, we find that TIM4 operates independently of lactadherin, which had been proposed to act as a bridging molecule. Of interest, TIM4-driven phagocytosis depends on the activation of integrins and involves stimulation of Src-family kinases and focal adhesion kinase, as well as the localized accumulation of phosphatidylinositol 3,4,5-trisphosphate. These mediators promote recruitment of the nucleotide-exchange factor Vav3, which in turn activates small Rho-family GTPases. Gene silencing or ablation experiments demonstrated that RhoA, Rac1, and Rac2 act synergistically to drive the remodeling of actin that underlies phagocytosis. Single-particle detection experiments demonstrated that TIM4 and β1 integrins associate upon receptor clustering. These findings support a model in which TIM4 engages integrins as coreceptors to evoke the signal transduction needed to internalize PtdSer-bearing targets such as apoptotic cells.
Collapse
Affiliation(s)
- Ronald S Flannagan
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON M5C 1N8, Canada
| | | | | | | | | |
Collapse
|
44
|
Maugeri N, Rovere-Querini P, Baldini M, Baldissera E, Sabbadini MG, Bianchi ME, Manfredi AA. Oxidative stress elicits platelet/leukocyte inflammatory interactions via HMGB1: a candidate for microvessel injury in sytemic sclerosis. Antioxid Redox Signal 2014; 20:1060-74. [PMID: 24070090 DOI: 10.1089/ars.2013.5298] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIMS An abnormal generation of reactive oxygen species (ROS) is thought to contribute to systemic sclerosis (SSc), fostering autoimmunity, fibrosis, and vascular inflammation. The function of the prototypic damage-associated molecular pattern, high mobility group box 1 (HMGB1), depends on its redox status. Here we investigate whether oxidative stress regulates the cross-talk between leukocytes and platelets via HMGB1, thus contributing to vessel inflammation in SSc. RESULTS The oxidation of HMGB1 amplified its ability to activate neutrophils, as detected assessing the redistribution of primary granule molecules and the transactivation of the β2 integrin chain CD18. Activated platelets are a source of bioactive HMGB1 and via P-selectin stimulated neutrophils to generate ROS. Oxidized extracellular HMGB1, soluble or associated to platelet membrane or to platelet-derived microparticles (PDμPs), further increased leukocyte activation. Leukocyte activation abated in the presence of inhibitors of HMGB1 or of catalase, which catalyzes the dismutation of hydrogen peroxide into water and molecular oxygen. The redistribution of the content of primary granules and the transactivation of β2 integrins characterized blood leukocytes of SSc patients and membrane HMGB1 was significantly higher in patients with pulmonary hypertension or with diffuse SSc. HMGB1(+) microparticles (μPs) purified from SSc patients, but not HMGB1(-) μPs purified from control subjects, activated in vitro healthy neutrophils, and HMGB1 inhibitors reversed the effects of μPs. INNOVATION AND CONCLUSION ROS dramatically increase the ability of extracellular HMGB1 to activate blood leukocytes. This event might contribute to maintain the microvascular injury of patients with SSc.
Collapse
Affiliation(s)
- Norma Maugeri
- 1 Division of Regenerative Medicine, Stem Cells and Gene Therapy, and Department of Medicine, San Raffaele Scientific Institute , Milano, Italy
| | | | | | | | | | | | | |
Collapse
|
45
|
Lutheran/basal cell adhesion molecule accelerates progression of crescentic glomerulonephritis in mice. Kidney Int 2014; 85:1123-36. [PMID: 24429403 PMCID: PMC4008878 DOI: 10.1038/ki.2013.522] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 10/17/2013] [Accepted: 10/24/2013] [Indexed: 12/14/2022]
Abstract
Migration of circulating leukocytes from the vasculature into the surrounding tissue is an important component of the inflammatory response. Among the cell surface molecules identified as contributing to leukocyte extravasation is VCAM-1, expressed on activated vascular endothelium, which participates in all stages of leukocyte–endothelial interaction by binding to leukocyte surface expressed integrin VLA-4. However, not all VLA-4-mediated events can be linked to VCAM-1. A novel interaction between VLA-4 and endothelial Lutheran (Lu) blood group antigens and basal cell adhesion molecule (BCAM) proteins has been recently shown, suggesting that Lu/BCAM may have a role in leukocyte recruitments in inflamed tissues. Here, we assessed the participation of Lu/BCAM in the immunopathogenesis of crescentic glomerulonephritis. High expression of Lu/BCAM in glomeruli of mice with rapidly progressive glomerulonephritis suggests a potential role for the local expression of Lu/BCAM in nephritogenic recruitment of leukocytes. Genetic deficiency of Lu/BCAM attenuated glomerular accumulation of T cells and macrophages, crescent formation, and proteinuria, correlating with reduced fibrin and platelet deposition in glomeruli. Furthermore, we found a pro-adhesive interaction between human monocyte α4β1 integrin and Lu/BCAM proteins. Thus, Lu/BCAM may have a critical role in facilitating the accumulation of monocytes and macrophages, thereby exacerbating renal injury.
Collapse
|
46
|
Stephen J, Emerson B, Fox KAA, Dransfield I. The Uncoupling of Monocyte–Platelet Interactions from the Induction of Proinflammatory Signaling in Monocytes. THE JOURNAL OF IMMUNOLOGY 2013; 191:5677-83. [DOI: 10.4049/jimmunol.1301250] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
47
|
Rondina MT, Weyrich AS, Zimmerman GA. Platelets as cellular effectors of inflammation in vascular diseases. Circ Res 2013; 112:1506-19. [PMID: 23704217 DOI: 10.1161/circresaha.113.300512] [Citation(s) in RCA: 235] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Platelets are chief effector cells in hemostasis. In addition, they are multifaceted inflammatory cells with functions that span the continuum from innate immune responses to adaptive immunity. Activated platelets have key thromboinflammatory activities in a variety of vascular disorders and vasculopathies. Recently identified inflammatory and immune activities provide insights into the biology of these versatile blood cells that are directly relevant to human vascular diseases.
Collapse
Affiliation(s)
- Matthew T Rondina
- Department of Medicine and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | | |
Collapse
|
48
|
Page C, Pitchford S. Neutrophil and platelet complexes and their relevance to neutrophil recruitment and activation. Int Immunopharmacol 2013; 17:1176-84. [PMID: 23810443 DOI: 10.1016/j.intimp.2013.06.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Revised: 09/20/2012] [Accepted: 06/09/2013] [Indexed: 12/29/2022]
Abstract
The manifestation of platelet 'satallitism' around neutrophils in whole blood is a long acknowledged phenomenon [1]. Circulating platelet-neutrophil complexes (PNC) occur in a diverse range of inflammatory disorders and infections that affect numerous organs of the body. Animal models have revealed that the formation of PNC is required for the recruitment of neutrophils to inflamed tissue, since platelets 'prime' neutrophils for efficient adhesion to vascular endothelium via the up-regulation of integrins and enhanced responsiveness to chemokines (Fig. 1). Perhaps surprisingly, the surface contact between platelets and neutrophils additionally enhances other neutrophil functions, such as chemotaxis that is required for migration into tissues, trans-cellular production of eicosanoids, phagocytosis and trapping of pathogens, increased respiratory burst leading to the production of reactive oxygen species (ROS), and modulation of neutrophil apoptosis (Fig. 1). Platelet P-selectin appears to have a particular role in enhancing the majority of these activities, and the influence of platelet P-selectin is not therefore confined to the initial rolling events in the process of neutrophil extravasation.
Collapse
Affiliation(s)
- Clive Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, Kings College London, London SE1 9NH, UK
| | | |
Collapse
|
49
|
Kim S, Cipolla L, Guidetti G, Okigaki M, Jin J, Torti M, Kunapuli SP. Distinct role of Pyk2 in mediating thromboxane generation downstream of both G12/13 and integrin αIIbβ3 in platelets. J Biol Chem 2013; 288:18194-203. [PMID: 23640884 DOI: 10.1074/jbc.m113.461087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Proline-rich tyrosine kinase 2 (Pyk2) is activated by various agonists in platelets. We evaluated the signaling mechanism and the functional role of Pyk2 in platelets by using pharmacological inhibitors and Pyk2-deficient platelets. We found that platelet aggregation and secretion in response to 2-methylthio-ADP (2-MeSADP) and AYPGKF were diminished in the presence of Pyk2 inhibitors or in Pyk2-deficient platelets, suggesting that Pyk2 plays a positive regulatory role in platelet functional responses. It has been shown that ADP-, but not thrombin-induced thromboxane (TxA2) generation depends on integrin signaling. Unlike ADP, thrombin activates G12/13 pathways, and G12/13 pathways can substitute for integrin signaling for TxA2 generation. We found that Pyk2 was activated downstream of both G12/13 and integrin-mediated pathways, and both 2-MeSADP- and AYPGKF-induced TxA2 generation was significantly diminished in Pyk2-deficient platelets. In addition, TxA2 generation induced by co-stimulation of Gi and Gz pathways, which is dependent on integrin signaling, was inhibited by blocking Pyk2. Furthermore, inhibition of 2-MeSADP-induced TxA2 generation by fibrinogen receptor antagonist was not rescued by co-stimulation of G12/13 pathways in the presence of Pyk2 inhibitor. We conclude that Pyk2 is a common signaling effector downstream of both G12/13 and integrin αIIbβ3 signaling, which contributes to thromboxane generation.
Collapse
Affiliation(s)
- Soochong Kim
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Herter J, Zarbock A. Integrin Regulation during Leukocyte Recruitment. THE JOURNAL OF IMMUNOLOGY 2013; 190:4451-7. [DOI: 10.4049/jimmunol.1203179] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|