1
|
Frelinger AL. Flow Cytometry and Platelets. Clin Lab Med 2024; 44:511-526. [PMID: 39089755 DOI: 10.1016/j.cll.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Clinical assessment of platelet activation by flow cytometry is useful in the characterization and diagnosis of platelet-specific disorders and as a measure of risk for thrombosis or bleeding. Platelets circulate in a resting, "unactivated" state, but when activated they undergo alterations in surface glycoprotein function and/or expression level, exposure of granule membrane proteins, and exposure of procoagulant phospholipids. Flow cytometry provides the means to detect these changes and, unlike other platelet tests, is appropriate for measuring platelet function in samples from patients with low platelet counts. The present review will focus on flow cytometric tests for platelet activation markers.
Collapse
Affiliation(s)
- Andrew L Frelinger
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115-5737, USA.
| |
Collapse
|
2
|
Kang YH, Varghese PM, Aiyan AA, Pondman K, Kishore U, Sim RB. Complement-Coagulation Cross-talk: Factor H-mediated regulation of the Complement Classical Pathway activation by fibrin clots. Front Immunol 2024; 15:1368852. [PMID: 38933264 PMCID: PMC11199686 DOI: 10.3389/fimmu.2024.1368852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/06/2024] [Indexed: 06/28/2024] Open
Abstract
The classical pathway of the complement system is activated by the binding of C1q in the C1 complex to the target activator, including immune complexes. Factor H is regarded as the key downregulatory protein of the complement alternative pathway. However, both C1q and factor H bind to target surfaces via charge distribution patterns. For a few targets, C1q and factor H compete for binding to common or overlapping sites. Factor H, therefore, can effectively regulate the classical pathway activation through such targets, in addition to its previously characterized role in the alternative pathway. Both C1q and factor H are known to recognize foreign or altered-self materials, e.g., bacteria, viruses, and apoptotic/necrotic cells. Clots, formed by the coagulation system, are an example of altered self. Factor H is present abundantly in platelets and is a well-known substrate for FXIIIa. Here, we investigated whether clots activate the complement classical pathway and whether this is regulated by factor H. We show here that both C1q and factor H bind to the fibrin formed in microtiter plates and the fibrin clots formed under in vitro physiological conditions. Both C1q and factor H become covalently bound to fibrin clots, and this is mediated via FXIIIa. We also show that fibrin clots activate the classical pathway of complement, as demonstrated by C4 consumption and membrane attack complex detection assays. Thus, factor H downregulates the activation of the classical pathway induced by fibrin clots. These results elucidate the intricate molecular mechanisms through which the complement and coagulation pathways intersect and have regulatory consequences.
Collapse
Affiliation(s)
- Yu-Hoi Kang
- Medical Research Council Immunochemistry Unit, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- MediMabBio Inc., Pangyo Business Growth Centre, Gyeonggi-do, Republic of Korea
| | - Praveen M. Varghese
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Ahmad Al Aiyan
- Department of Veterinary Medicine (CAVM), United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kirsten Pondman
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology & TechMed Centre, University of Twente, Enschede, Netherlands
| | - Uday Kishore
- Department of Veterinary Medicine (CAVM), United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Robert B. Sim
- Medical Research Council Immunochemistry Unit, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
3
|
Thom CS, Davenport P, Fazelinia H, Soule-Albridge E, Liu ZJ, Zhang H, Feldman HA, Ding H, Roof J, Spruce LA, Ischiropoulos H, Sola-Visner M. Quantitative label-free mass spectrometry reveals content and signaling differences between neonatal and adult platelets. J Thromb Haemost 2024; 22:1447-1462. [PMID: 38160730 PMCID: PMC11055671 DOI: 10.1016/j.jtha.2023.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Recent clinical studies have shown that transfusions of adult platelets increase morbidity and mortality in preterm infants. Neonatal platelets are hyporesponsive to agonist stimulation, and emerging evidence suggests developmental differences in platelet immune functions. OBJECTIVES This study was designed to compare the proteome and phosphoproteome of resting adult and neonatal platelets. METHODS We isolated resting umbilical cord blood-derived platelets from healthy full-term neonates (n = 8) and resting blood platelets from healthy adults (n = 6) and compared protein and phosphoprotein contents using data-independent acquisition mass spectrometry. RESULTS We identified 4770 platelet proteins with high confidence across all samples. Adult and neonatal platelets were clustered separately by principal component analysis. Adult platelets were significantly enriched in immunomodulatory proteins, including β2 microglobulin and CXCL12, whereas neonatal platelets were enriched in ribosomal components and proteins involved in metabolic activities. Adult platelets were enriched in phosphorylated GTPase regulatory enzymes and proteins participating in trafficking, which may help prime them for activation and degranulation. Neonatal platelets were enriched in phosphorylated proteins involved in insulin growth factor signaling. CONCLUSION Using label-free data-independent acquisition mass spectrometry, our findings expanded the known neonatal platelet proteome and identified important differences in protein content and phosphorylation between neonatal and adult platelets. These developmental differences suggested enhanced immune functions for adult platelets and presence of molecular machinery related to platelet activation. These findings are important to understanding mechanisms underlying key platelet functions as well as the harmful effects of adult platelet transfusions given to preterm infants.
Collapse
Affiliation(s)
- Christopher S Thom
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Patricia Davenport
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Hossein Fazelinia
- Proteomics Core, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Erin Soule-Albridge
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Zhi-Jian Liu
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Haorui Zhang
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Henry A Feldman
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA; Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Hua Ding
- Proteomics Core, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jennifer Roof
- Proteomics Core, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lynn A Spruce
- Proteomics Core, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Harry Ischiropoulos
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Martha Sola-Visner
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
4
|
Xie HG, Jiang LP, Tai T, Ji JZ, Mi QY. The Complement System and C4b-Binding Protein: A Focus on the Promise of C4BPα as a Biomarker to Predict Clopidogrel Resistance. Mol Diagn Ther 2024; 28:189-199. [PMID: 38261250 DOI: 10.1007/s40291-023-00691-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2023] [Indexed: 01/24/2024]
Abstract
The complement system plays a dual role in the body, either as a first-line defense barrier when balanced between activation and inhibition or as a potential driver of complement-associated injury or diseases when unbalanced or over-activated. C4b-binding protein (C4BP) was the first circulating complement regulatory protein identified and it functions as an important complement inhibitor. C4BP can suppress the over-activation of complement components and prevent the complement system from attacking the host cells through the binding of complement cleavage products C4b and C3b, working in concert as a cofactor for factor I in the degradation of C4b and C3b, and consequently preventing or reducing the assembly of C3 convertase and C5 convertase, respectively. C4BP, particularly C4BP α-chain (C4BPα), exerts its unique inhibitory effects on complement activation and opsonization, systemic inflammation, and platelet activation and aggregation. It has long been acknowledged that crosstalk or interplay exists between the complement system and platelets. Our unpublished preliminary data suggest that circulating C4BPα exerts its antiplatelet effects through inhibition of both complement activity levels and complement-induced platelet reactivity. Plasma C4BPα levels appear to be significantly higher in patients sensitive to, rather than resistant to, clopidogrel, and we suggest that a plasma C4BPα measurement could be used to predict clopidogrel resistance in the clinical settings.
Collapse
Affiliation(s)
- Hong-Guang Xie
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China.
| | - Li-Ping Jiang
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China
| | - Ting Tai
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China
| | - Jin-Zi Ji
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China
| | - Qiong-Yu Mi
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China
| |
Collapse
|
5
|
Andersson LI, Sjöström DJ, Quach HQ, Hägerström K, Hurler L, Kajdácsi E, Cervenak L, Prohászka Z, Toonen EJM, Mohlin C, Mollnes TE, Sandgren P, Tjernberg I, Nilsson PH. Storage of Transfusion Platelet Concentrates Is Associated with Complement Activation and Reduced Ability of Platelets to Respond to Protease-Activated Receptor-1 and Thromboxane A2 Receptor. Int J Mol Sci 2024; 25:1091. [PMID: 38256162 PMCID: PMC10816124 DOI: 10.3390/ijms25021091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Platelet activation and the complement system are mutually dependent. Here, we investigated the effects of storage time on complement activation and platelet function in routinely produced platelet concentrates. The platelet concentrates (n = 10) were stored at 22 °C for seven days and assessed daily for complement and platelet activation markers. Additionally, platelet function was analyzed in terms of their responsiveness to protease-activated receptor-1 (PAR-1) and thromboxane A2 receptor (TXA2R) activation and their capacity to adhere to collagen. Complement activation increased over the storage period for all analyzed markers, including the C1rs/C1-INH complex (fold change (FC) = 1.9; p < 0.001), MASP-1/C1-INH complex (FC = 2.0; p < 0.001), C4c (FC = 1.8, p < 0.001), C3bc (FC = 4.0; p < 0.01), and soluble C5b-9 (FC = 1.7, p < 0.001). Furthermore, the levels of soluble platelet activation markers increased in the concentrates over the seven-day period, including neutrophil-activating peptide-2 (FC = 2.5; p < 0.0001), transforming growth factor beta 1 (FC = 1.9; p < 0.001) and platelet factor 4 (FC = 2.1; p < 0.0001). The ability of platelets to respond to activation, as measured by surface expression of CD62P and CD63, decreased by 19% and 24% (p < 0.05) for PAR-1 and 69-72% (p < 0.05) for TXA2R activation, respectively, on Day 7 compared to Day 1. The extent of platelet binding to collagen was not significantly impaired during storage. In conclusion, we demonstrated that complement activation increased during the storage of platelets, and this correlated with increased platelet activation and a reduced ability of the platelets to respond to, primarily, TXA2R activation.
Collapse
Affiliation(s)
- Linnea I. Andersson
- Department of Chemistry and Biomedicine, Linnaeus University, 391 82 Kalmar, Sweden; (L.I.A.); (D.J.S.); (C.M.)
| | - Dick J. Sjöström
- Department of Chemistry and Biomedicine, Linnaeus University, 391 82 Kalmar, Sweden; (L.I.A.); (D.J.S.); (C.M.)
| | - Huy Quang Quach
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA;
| | - Kim Hägerström
- Department of Clinical Chemistry and Transfusion Medicine, Region Kalmar County, 391 85 Kalmar, Sweden; (K.H.); (I.T.)
| | - Lisa Hurler
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary; (L.H.); (E.K.); (L.C.); (Z.P.)
| | - Erika Kajdácsi
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary; (L.H.); (E.K.); (L.C.); (Z.P.)
| | - László Cervenak
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary; (L.H.); (E.K.); (L.C.); (Z.P.)
| | - Zoltán Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary; (L.H.); (E.K.); (L.C.); (Z.P.)
| | | | - Camilla Mohlin
- Department of Chemistry and Biomedicine, Linnaeus University, 391 82 Kalmar, Sweden; (L.I.A.); (D.J.S.); (C.M.)
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital, University of Oslo, 0372 Oslo, Norway;
- Research Laboratory, Nordland Hospital, 8005 Bodo, Norway
| | - Per Sandgren
- Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, 171 77 Huddinge, Sweden;
| | - Ivar Tjernberg
- Department of Clinical Chemistry and Transfusion Medicine, Region Kalmar County, 391 85 Kalmar, Sweden; (K.H.); (I.T.)
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping University, 581 83 Linköping, Sweden
| | - Per H. Nilsson
- Department of Chemistry and Biomedicine, Linnaeus University, 391 82 Kalmar, Sweden; (L.I.A.); (D.J.S.); (C.M.)
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, 391 82 Kalmar, Sweden
| |
Collapse
|
6
|
Davenport P, Soule-Albridge E, Sola-Visner M. Hemostatic and Immunologic Effects of Platelet Transfusions in Neonates. Clin Perinatol 2023; 50:793-803. [PMID: 37866848 DOI: 10.1016/j.clp.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Liberal platelet transfusions are associated with increased morbidity and mortality among preterm neonates, and it is now recognized that platelets are both hemostatic and immune cells. Neonatal and adult platelets are functionally distinct, and adult platelets have the potential to be more immuno-active. Preclinical studies suggest that platelet transfusions (from adult donors) can trigger dysregulated immune responses in neonates, which might mediate the increased morbidity and mortality observed in clinical studies. More research is needed to understand how neonatal and adult platelets differ in their immune functions and the consequences of these differences in the setting of neonatal platelet transfusions.
Collapse
Affiliation(s)
- Patricia Davenport
- Division of Newborn Medicine, Boston Children's Hospital, 300 Longwood Avenue, Enders 954, Boston, MA 02115, USA.
| | - Erin Soule-Albridge
- Division of Newborn Medicine, Boston Children's Hospital, 300 Longwood Avenue, Enders 950.5, Boston, MA 02115, USA
| | - Martha Sola-Visner
- Division of Newborn Medicine, Boston Children's Hospital, 300 Longwood Avenue, Enders 961, Boston, MA 02115, USA
| |
Collapse
|
7
|
Xing Y, Zhang D, Fang L, Wang J, Liu C, Wu D, Liu X, Wang X, Min W. Complement in Human Brain Health: Potential of Dietary Food in Relation to Neurodegenerative Diseases. Foods 2023; 12:3580. [PMID: 37835232 PMCID: PMC10572247 DOI: 10.3390/foods12193580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
The complement pathway is a major component of the innate immune system, which is critical for recognizing and clearing pathogens that rapidly react to defend the body against external pathogens. Many components of this pathway are expressed throughout the brain and play a beneficial role in synaptic pruning in the developing central nervous system (CNS). However, excessive complement-mediated synaptic pruning in the aging or injured brain may play a contributing role in a wide range of neurodegenerative diseases. Complement Component 1q (C1q), an initiating recognition molecule of the classical complement pathway, can interact with a variety of ligands and perform a range of functions in physiological and pathophysiological conditions of the CNS. This review considers the function and immunomodulatory mechanisms of C1q; the emerging role of C1q on synaptic pruning in developing, aging, or pathological CNS; the relevance of C1q; the complement pathway to neurodegenerative diseases; and, finally, it summarizes the foods with beneficial effects in neurodegenerative diseases via C1q and complement pathway and highlights the need for further research to clarify these roles. This paper aims to provide references for the subsequent study of food functions related to C1q, complement, neurodegenerative diseases, and human health.
Collapse
Affiliation(s)
- Yihang Xing
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.X.); (D.Z.); (L.F.); (J.W.); (C.L.); (D.W.); (X.L.)
| | - Dingwen Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.X.); (D.Z.); (L.F.); (J.W.); (C.L.); (D.W.); (X.L.)
| | - Li Fang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.X.); (D.Z.); (L.F.); (J.W.); (C.L.); (D.W.); (X.L.)
| | - Ji Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.X.); (D.Z.); (L.F.); (J.W.); (C.L.); (D.W.); (X.L.)
| | - Chunlei Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.X.); (D.Z.); (L.F.); (J.W.); (C.L.); (D.W.); (X.L.)
| | - Dan Wu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.X.); (D.Z.); (L.F.); (J.W.); (C.L.); (D.W.); (X.L.)
| | - Xiaoting Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.X.); (D.Z.); (L.F.); (J.W.); (C.L.); (D.W.); (X.L.)
| | - Xiyan Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.X.); (D.Z.); (L.F.); (J.W.); (C.L.); (D.W.); (X.L.)
| | - Weihong Min
- College of Food and Health, Zhejiang A&F University, Hangzhou 311300, China
| |
Collapse
|
8
|
Thom CS, Davenport P, Fazelinia H, Liu ZJ, Zhang H, Ding H, Roof J, Spruce LA, Ischiropoulos H, Sola-Visner M. Phosphoproteomics reveals content and signaling differences between neonatal and adult platelets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.13.557268. [PMID: 37745418 PMCID: PMC10515911 DOI: 10.1101/2023.09.13.557268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Background and Objective Recent clinical studies have shown that transfusions of adult platelets increase morbidity and mortality in preterm infants. Neonatal platelets are hyporesponsive to agonist stimulation, and emerging evidence suggests developmental differences in platelet immune functions. This study was designed to compare the proteome and phosphoproteome of resting adult and neonatal platelets. Methods We isolated resting umbilical cord blood-derived platelets from healthy full term neonates (n=9) and resting blood platelets from healthy adults (n=7), and compared protein and phosphoprotein contents using data independent acquisition mass spectrometry. Results We identified 4745 platelet proteins with high confidence across all samples. Adult and neonatal platelets clustered separately by principal component analysis. Adult platelets were significantly enriched for immunomodulatory proteins, including β2 microglobulin and CXCL12, whereas neonatal platelets were enriched for ribosomal components and proteins involved in metabolic activities. Adult platelets were enriched for phosphorylated GTPase regulatory enzymes and proteins participating in trafficking, which may help prime them for activation and degranulation. Neonatal platelets were enriched for phosphorylated proteins involved in insulin growth factor signaling. Conclusions Using state-of-the-art mass spectrometry, our findings expanded the known neonatal platelet proteome and identified important differences in protein content and phosphorylation compared with adult platelets. These developmental differences suggested enhanced immune functions for adult platelets and presence of a molecular machinery related to platelet activation. These findings are important to understanding mechanisms underlying key platelet functions as well as the harmful effects of adult platelet transfusions given to preterm infants.
Collapse
Affiliation(s)
- Christopher S Thom
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patricia Davenport
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Hossein Fazelinia
- Proteomics Core, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zhi-Jian Liu
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Haorui Zhang
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Hua Ding
- Proteomics Core, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jennifer Roof
- Proteomics Core, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lynn A Spruce
- Proteomics Core, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Harry Ischiropoulos
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA
| | - Martha Sola-Visner
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
9
|
Santiago VF, Dombrowski JG, Kawahara R, Rosa-Fernandes L, Mule SN, Murillo O, Santana TV, Coutinho JVP, Macedo-da-Silva J, Lazari LC, Peixoto EPM, Ramirez MI, Larsen MR, Marinho CRF, Palmisano G. Complement System Activation Is a Plasma Biomarker Signature during Malaria in Pregnancy. Genes (Basel) 2023; 14:1624. [PMID: 37628675 PMCID: PMC10454407 DOI: 10.3390/genes14081624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Malaria in pregnancy (MiP) is a public health problem in malaria-endemic areas, contributing to detrimental outcomes for both mother and fetus. Primigravida and second-time mothers are most affected by severe anemia complications and babies with low birth weight compared to multigravida women. Infected erythrocytes (IE) reach the placenta, activating the immune response by placental monocyte infiltration and inflammation. However, specific markers of MiP result in poor outcomes, such as low birth weight, and intrauterine growth restriction for babies and maternal anemia in women infected with Plasmodium falciparum are limited. In this study, we identified the plasma proteome signature of a mouse model infected with Plasmodium berghei ANKA and pregnant women infected with Plasmodium falciparum infection using quantitative mass spectrometry-based proteomics. A total of 279 and 249 proteins were quantified in murine and human plasma samples, of which 28% and 30% were regulated proteins, respectively. Most of the regulated proteins in both organisms are involved in complement system activation during malaria in pregnancy. CBA anaphylatoxin assay confirmed the complement system activation by the increase in C3a and C4a anaphylatoxins in the infected plasma compared to non-infected plasma. Moreover, correlation analysis showed the association between complement system activation and reduced head circumference in newborns from Pf-infected mothers. The data obtained in this study highlight the correlation between the complement system and immune and newborn outcomes resulting from malaria in pregnancy.
Collapse
Affiliation(s)
- Veronica Feijoli Santiago
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Jamille Gregorio Dombrowski
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Rebeca Kawahara
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
- Analytical Glycoimmunology Group, Department of Molecular Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Livia Rosa-Fernandes
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Simon Ngao Mule
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Oscar Murillo
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center Science at Tyler, Tyler, TX 75708, USA
| | - Thais Viggiani Santana
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Joao Victor Paccini Coutinho
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Janaina Macedo-da-Silva
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Lucas Cardoso Lazari
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Erika Paula Machado Peixoto
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Marcel Ivan Ramirez
- Cell Biology Laboratory, Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, Brazil
| | - Martin R. Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | | | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
- Analytical Glycoimmunology Group, Department of Molecular Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| |
Collapse
|
10
|
Cavalcante JS, de Almeida DEG, Santos-Filho NA, Sartim MA, de Almeida Baldo A, Brasileiro L, Albuquerque PL, Oliveira SS, Sachett JAG, Monteiro WM, Ferreira RS. Crosstalk of Inflammation and Coagulation in Bothrops Snakebite Envenoming: Endogenous Signaling Pathways and Pathophysiology. Int J Mol Sci 2023; 24:11508. [PMID: 37511277 PMCID: PMC10380640 DOI: 10.3390/ijms241411508] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/26/2023] [Accepted: 06/05/2023] [Indexed: 07/30/2023] Open
Abstract
Snakebite envenoming represents a major health problem in tropical and subtropical countries. Considering the elevated number of accidents and high morbidity and mortality rates, the World Health Organization reclassified this disease to category A of neglected diseases. In Latin America, Bothrops genus snakes are mainly responsible for snakebites in humans, whose pathophysiology is characterized by local and systemic inflammatory and degradative processes, triggering prothrombotic and hemorrhagic events, which lead to various complications, organ damage, tissue loss, amputations, and death. The activation of the multicellular blood system, hemostatic alterations, and activation of the inflammatory response are all well-documented in Bothrops envenomings. However, the interface between inflammation and coagulation is still a neglected issue in the toxinology field. Thromboinflammatory pathways can play a significant role in some of the major complications of snakebite envenoming, such as stroke, venous thromboembolism, and acute kidney injury. In addition to exacerbating inflammation and cell interactions that trigger vaso-occlusion, ischemia-reperfusion processes, and, eventually, organic damage and necrosis. In this review, we discuss the role of inflammatory pathways in modulating coagulation and inducing platelet and leukocyte activation, as well as the inflammatory production mediators and induction of innate immune responses, among other mechanisms that are altered by Bothrops venoms.
Collapse
Affiliation(s)
- Joeliton S Cavalcante
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP-Univ Estadual Paulista), Botucatu 18618-687, São Paulo, Brazil
| | - Denis Emanuel Garcia de Almeida
- Department of Bioprocess and Biotechnology, School of Agriculture, Agronomic Sciences School, São Paulo State University (UNESP-Univ Estadual Paulista), Botucatu 18618-687, São Paulo, Brazil
| | - Norival A Santos-Filho
- Institute of Chemistry, São Paulo State University (UNESP-Univ Estadual Paulista), Araraquara 14800-900, São Paulo, Brazil
| | - Marco Aurélio Sartim
- Laboratory of Bioprospection, University Nilton Lins, Manaus 69058-030, Amazonas, Brazil
- Research & Development Department, Nilton Lins Foundation, Manaus 69058-030, Amazonas, Brazil
- Graduate Program in Tropical Medicine, Department of Research at Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Amazonas State University, Manaus 69850-000, Amazonas, Brazil
| | - Amanda de Almeida Baldo
- Institute of Biosciences, São Paulo State University (UNESP-Univ Estadual Paulista), Botucatu 18618-687, São Paulo, Brazil
| | - Lisele Brasileiro
- Graduate Program in Tropical Medicine, Department of Research at Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Amazonas State University, Manaus 69850-000, Amazonas, Brazil
| | - Polianna L Albuquerque
- Toxicological Information and Assistance Center, Instituto Doutor Jose Frota Hospital, Fortaleza 60025-061, Ceará, Brazil
- Faculty of Medicine, University of Fortaleza, Fortaleza 60430-140, Ceará, Brazil
| | - Sâmella S Oliveira
- Research Management, Hospital Foundation of Hematology and Hemotherapy of Amazonas, Manaus 69050-001, Amazonas, Brazil
| | - Jacqueline Almeida Gonçalves Sachett
- Research & Development Department, Nilton Lins Foundation, Manaus 69058-030, Amazonas, Brazil
- Graduate Program in Tropical Medicine, Department of Research at Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Amazonas State University, Manaus 69850-000, Amazonas, Brazil
| | - Wuelton Marcelo Monteiro
- Research & Development Department, Nilton Lins Foundation, Manaus 69058-030, Amazonas, Brazil
- Graduate Program in Tropical Medicine, Department of Research at Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Amazonas State University, Manaus 69850-000, Amazonas, Brazil
| | - Rui Seabra Ferreira
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP-Univ Estadual Paulista), Botucatu 18618-687, São Paulo, Brazil
- Center for Translational Science and Development of Biopharmaceuticals FAPESP/CEVAP-UNESP, Botucatu 18610-307, São Paulo, Brazil
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP-Univ Estadual Paulista), Botucatu 18610-307, São Paulo, Brazil
| |
Collapse
|
11
|
Abstract
The complement and hemostatic systems are complex systems, and both involve enzymatic cascades, regulators, and cell components-platelets, endothelial cells, and immune cells. The two systems are ancestrally related and are defense mechanisms that limit infection by pathogens and halt bleeding at the site of vascular injury. Recent research has uncovered multiple functional interactions between complement and hemostasis. On one side, there are proteins considered as complement factors that activate hemostasis, and on the other side, there are coagulation proteins that modulate complement. In addition, complement and coagulation and their regulatory proteins strongly interact each other to modulate endothelial, platelet and leukocyte function and phenotype, creating a potentially devastating amplifying system that must be closely regulated to avoid unwanted damage and\or disseminated thrombosis. In view of its ability to amplify all complement activity through the C3b-dependent amplification loop, the alternative pathway of complement may play a crucial role in this context. In this review, we will focus on available and emerging evidence on the role of the alternative pathway of complement in regulating hemostasis and vice-versa, and on how dysregulation of either system can lead to severe thromboinflammatory events.
Collapse
Affiliation(s)
- Marina Noris
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Miriam Galbusera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
12
|
Stefanski AL, Nitschke E, Dörner T. Thromboinflammation: Dynamik physiologischer und pathologischer
Wechselwirkungen von Entzündung und Koagulation. AKTUEL RHEUMATOL 2022. [DOI: 10.1055/a-1947-5200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
ZusammenfassungDas konzertante Zusammenspiel zwischen endothelialer Dysfuntion, aktivierten
Thrombozyten und anderen Immunzellen sowie simultaner Komplementaktivierung
führt zur Aktivierung und gegenseitigen Verstärkung sowohl der
Immunantwort als auch der Gerinnungskaskade. Durch die unkontrollierte Fortdauer
dieser physiologischen Mechanismen kann der pathologische Prozess der
Thromboinflammation induziert werden. In dieser Übersichtsarbeit fassen
wir grundlegende Mechanismen zusammen, die zur Thromboinflammation als ein
Auslöser von venösen Thromboembolien führen.
Collapse
Affiliation(s)
- Ana-Luisa Stefanski
- Department of Rheumatology and Clinical Immunology, Charité
Universitätsmedizin Berlin Campus Charité Mitte, Berlin,
Germany
- Autoimmunity, DRFZ, Berlin, Germany
| | - Eduard Nitschke
- Department of Rheumatology and Clinical Immunology, Charité
Universitätsmedizin Berlin Campus Charité Mitte, Berlin,
Germany
- Autoimmunity, DRFZ, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité
Universitätsmedizin Berlin Campus Charité Mitte, Berlin,
Germany
- Autoimmunity, DRFZ, Berlin, Germany
| |
Collapse
|
13
|
Indications that the Antimycotic Drug Amphotericin B Enhances the Impact of Platelets on Aspergillus. Antimicrob Agents Chemother 2022; 66:e0068122. [PMID: 36190233 PMCID: PMC9578436 DOI: 10.1128/aac.00681-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Platelets are currently thought to harbor antimicrobial functions and might therefore play a crucial role in infections, e.g., those caused by Aspergillus or mucormycetes. The incidence of invasive fungal infections is increasing, particularly during the coronavirus disease 2019 (COVID-19) pandemic, and such infections continue to be life-threatening in immunocompromised patients. For this reason, the interaction of antimycotics with platelets is a key issue to evaluate modern therapeutic regimens. Amphotericin B (AmB) is widely used for the therapy of invasive fungal infections either as deoxycholate (AmB-D) or as a liposomal formulation (L-AmB). We showed that AmB strongly activates platelets within a few minutes. AmB concentrations commonly measured in the blood of patients were sufficient to stimulate platelets, indicating that this effect is highly relevant in vivo. The stimulating effect was corroborated by a broad spectrum of platelet activation parameters, including degranulation, aggregation, budding of microparticles, morphological changes, and enhanced adherence to fungal hyphae. Comparison between the deoxycholate and the liposomal formulation excluded the possibility that the liposomal part of L-Amb is responsible for these effects, as no difference was visible. The induction of platelet activation and alteration by L-AmB resulted in the activation of other parts of innate immunity, such as stimulation of the complement cascade and interaction with granulocytes. These mechanisms might substantially fuel the antifungal immune reaction in invasive mycoses. On the other hand, thrombosis and excessive inflammatory processes might occur via these mechanisms. Furthermore, the viability of L-AmB-activated platelets was consequently decreased, a process that might contribute to thrombocytopenia in patients.
Collapse
|
14
|
Xing Z, Wang Y, Gong K, Chen Y. Plasma C4 level was associated with mortality, cardiovascular and cerebrovascular complications in hemodialysis patients. BMC Nephrol 2022; 23:232. [PMID: 35768780 PMCID: PMC9245318 DOI: 10.1186/s12882-022-02829-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/19/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients on maintenance hemodialysis (HD) exhibit a high risk of death, cardiovascular and cerebrovascular diseases (CCDs). Previous studies indicated complement activation associated with the increased risk of cardiovascular diseases in HD patients. This study aimed to explore whether the critical complement factors were associated with the adverse outcomes in HD patients. METHODS A total of 108 HD patients were included and followed up for 52 months. The baseline clinical characteristics and plasma C3c, C1q, CFH, CFB, C4, MAC, C5a, C3a and MBL were measured. The three endpoints were death, cardiovascular and cerebrovascular events (CCEs) and the composition of them. Univariate and multivariate Cox regression identified factors associated with the three endpoints respectively. X-tile analyses determined the optimal cut-off values for high risks. Restricted cubic spline plots illustrated the dose-response relationships. Correlations between the complement factors and risk factors for CCDs were analyzed. RESULTS Baseline plasma C4 was finally selected by univariate and multivariate Cox regression analyses for three endpoints, including all-cause mortality, CCEs and the composition of them. When baseline plasma C4 exceeded 0.47 (P = 0.001) or 0.44 (P = 0.018) g/L respectively, the risks for death or achieving the composite endpoint enhanced significantly. The relationships of C4 and HR for the three endpoints showed a positive linear trend. Plasma C4 had prominent correlations with blood TG (r = 0.62, P < 0.001) and HDL (r = -0.38, P < 0.001). CONCLUSIONS A higher baseline plasma C4 level was significantly associated with the future incidence of decease, CCEs and either of them. Plasma C4 level correlated with blood TG and HDL.
Collapse
Affiliation(s)
- Zheyu Xing
- Renal Division, Peking University First Hospital, Beijing, China.,Institute of Nephrology, Peking University, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Yaqin Wang
- Renal Division, Peking University First Hospital, Beijing, China.,Institute of Nephrology, Peking University, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Kunjing Gong
- Renal Division, Peking University First Hospital, Beijing, China.,Institute of Nephrology, Peking University, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Yuqing Chen
- Renal Division, Peking University First Hospital, Beijing, China. .,Institute of Nephrology, Peking University, Beijing, China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China. .,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China.
| |
Collapse
|
15
|
Linge CP, Jern A, Tydén H, Gullstrand B, Yan H, Welinder C, Kahn R, Jönssen A, Semple JW, Bengtsson AA. Enrichment of complement, immunoglobulins and autoantibody targets in the proteome of platelets from patients with Systemic Lupus Erythematosus (SLE). Thromb Haemost 2022; 122:1486-1501. [PMID: 35419777 PMCID: PMC9420555 DOI: 10.1055/a-1825-2915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background
Systemic lupus erythematosus (SLE) is a complex disease characterized by autoimmunity toward apoptotic cells, excessive amounts of circulating immune complexes, and complement activation. A decreased platelet size has been observed in SLE and their nonhemostatic functions may play an active role in the disease. The main objective of this study was to find clues that could explain their decreased size and functional role, analyzing the entire platelet proteome.
Methods
Platelets were isolated from 23 patients with SLE. The five individuals with the highest and lowest average platelet forward scatter were selected for further analysis. Platelet protein content was analyzed using liquid chromatography with tandem mass spectrometry (LC-MS/MS) and compared with platelets from five healthy controls. Data are available via ProteomeXchange with identifier PXD031202.
Results
Out of 2,572 proteins identified, 396 had significantly different levels (ANOVA
q
-value ≤ 0.01). Forty proteins, including immunoglobulin-, complement- and phosphatidylserine-binding proteins had higher abundance in platelets from SLE patients, largely independent of size (fold difference of ≥1.5 and a
t
-test
p
-value of ≤0.05 as cut-off). Functional characterization revealed increased degranulation and skewed hemostatic balance in platelets from SLE patients. In the SLE proteome, immunoglobulin proteins were negatively correlated to serum complement C3 and C4 and the highest relative levels were detected in platelets of normal size.
Conclusion
Platelets from SLE patients shared a specific protein profile, including immunoglobulins, complement proteins, and autoantigens, largely independent of the platelet size and in agreement with an integrated role for platelets in SLE.
Collapse
Affiliation(s)
- Carl Petrus Linge
- Department of Clinical Sciences Lund, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Andreas Jern
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Helena Tydén
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Birgitta Gullstrand
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Hong Yan
- BioMS, Swedish National Infrastructure for Biological Mass Spectrometry, Lund, Sweden
| | - Charlotte Welinder
- Department of Clinical Sciences Lund, Lund University Department of Oncology and Pathology, Lund, Sweden
| | - Robin Kahn
- Wallenberg Center for Molecular Medicin, Lund University Faculty of Medicine, Lund, Sweden.,Paediatrics, Lund University Faculty of Medicine, Lund, Sweden
| | - Andreas Jönssen
- Department of Clinical Sciences Lund, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - John W Semple
- Transfusion Medicine, Lunds Universitet, Lund, Sweden
| | - Anders A Bengtsson
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| |
Collapse
|
16
|
Jurasz P, Ignjatovic V, Lordkipanidzé M. Editorial: Established and Novel Roles of Platelets in Health and Disease. Front Cardiovasc Med 2022; 9:835615. [PMID: 35174235 PMCID: PMC8841832 DOI: 10.3389/fcvm.2022.835615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Vera Ignjatovic
- Department of Haematology Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
- *Correspondence: Marie Lordkipanidzé
| |
Collapse
|
17
|
Ostrycharz E, Hukowska-Szematowicz B. New Insights into the Role of the Complement System in Human Viral Diseases. Biomolecules 2022; 12:226. [PMID: 35204727 PMCID: PMC8961555 DOI: 10.3390/biom12020226] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/23/2022] [Accepted: 01/27/2022] [Indexed: 01/27/2023] Open
Abstract
The complement system (CS) is part of the human immune system, consisting of more than 30 proteins that play a vital role in the protection against various pathogens and diseases, including viral diseases. Activated via three pathways, the classical pathway (CP), the lectin pathway (LP), and the alternative pathway (AP), the complement system leads to the formation of a membrane attack complex (MAC) that disrupts the membrane of target cells, leading to cell lysis and death. Due to the increasing number of reports on its role in viral diseases, which may have implications for research on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), this review aims to highlight significant progress in understanding and defining the role of the complement system in four groups of diseases of viral etiology: (1) respiratory diseases; (2) acute liver failure (ALF); (3) disseminated intravascular coagulation (DIC); and (4) vector-borne diseases (VBDs). Some of these diseases already present a serious global health problem, while others are a matter of concern and require the collaboration of relevant national services and scientists with the World Health Organization (WHO) to avoid their spread.
Collapse
Affiliation(s)
- Ewa Ostrycharz
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland;
- Doctoral School of the University of Szczecin, University of Szczecin, 71-412 Szczecin, Poland
- Molecular Biology and Biotechnology Center, University of Szczecin, 71-412 Szczecin, Poland
| | - Beata Hukowska-Szematowicz
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland;
- Molecular Biology and Biotechnology Center, University of Szczecin, 71-412 Szczecin, Poland
| |
Collapse
|
18
|
Abstract
Classically, platelets have been described as the cellular blood component that mediates hemostasis and thrombosis. This important platelet function has received significant research attention for >150 years. The immune cell functions of platelets are much less appreciated. Platelets interact with and activate cells of all branches of immunity in response to pathogen exposures and infection, as well as in response to sterile tissue injury. In this review, we focus on innate immune mechanisms of platelet activation, platelet interactions with innate immune cells, as well as the intersection of platelets and adaptive immunity. The immune potential of platelets is dependent in part on their megakaryocyte precursor providing them with the molecular composition to be first responders and immune sentinels in initiating and orchestrating coordinated pathogen immune responses. There is emerging evidence that extramedullary megakaryocytes may be immune differentiated compared with bone marrow megakaryocytes, but the physiological relevance of immunophenotypic differences are just beginning to be explored. These concepts are also discussed in this review. The immune functions of the megakaryocyte/platelet lineage have likely evolved to coordinate the need to repair a vascular breach with the simultaneous need to induce an immune response that may limit pathogen invasion once the blood is exposed to an external environment.
Collapse
Affiliation(s)
- Milka Koupenova
- Department of Medicine, Division of Cardiovascular Medicine, University of Massachusetts Chan Medical School, 368 Plantation Street, Worcester, MA 01605
| | - Alison Livada
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642
| | - Craig N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY 14642
- Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
19
|
Abstract
Tumorigenesis has long been linked to the evasion of the immune system and the uncontrolled proliferation of transformed cells. The complement system, a major arm of innate immunity, is a key factor in the progression of cancer because many of its components have critical regulatory roles in the tumor microenvironment. For example, complement anaphylatoxins directly and indirectly inhibit antitumor T-cell responses in primary and metastatic sites, enhance proliferation of tumor cells, and promote metastasis and tumor angiogenesis. Many recent studies have provided evidence that cancer is able to hijack the immunoregulatory components of the complement system which fundamentally are tasked with protecting the body against abnormal cells and pathogens. Indeed, recent evidence shows that many types of cancer use C1q receptors (C1qRs) to promote tumor growth and progression. More importantly, most cancer cells express both C1q and its major receptors (gC1qR and cC1qR) on their surface which are essential for cell proliferation and survival. In this review, we discuss the ability of cancer to control and manipulate the complement system in the tumor microenvironment and identify possible therapeutic targets, including C1q and gC1qR.
Collapse
Affiliation(s)
- Danyaal Ain
- The Department of Medicine, Stony Brook University, 100 Nicholls Road, Stony Brook, NY 11794-8161, USA
| | - Talha Shaikh
- The Department of Medicine, Stony Brook University, 100 Nicholls Road, Stony Brook, NY 11794-8161, USA
| | - Samantha Manimala
- The Department of Medicine, Stony Brook University, 100 Nicholls Road, Stony Brook, NY 11794-8161, USA
| | - Berhane Ghebrehiwet
- The Department of Medicine, Stony Brook University, 100 Nicholls Road, Stony Brook, NY 11794-8161, USA
| |
Collapse
|
20
|
Mollnes TE, Storm BS, Brekke OL, Nilsson PH, Lambris JD. Application of the C3 inhibitor compstatin in a human whole blood model designed for complement research - 20 years of experience and future perspectives. Semin Immunol 2022; 59:101604. [PMID: 35570131 DOI: 10.1016/j.smim.2022.101604] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 04/23/2022] [Indexed: 01/15/2023]
Abstract
The complex molecular and cellular biological systems that maintain host homeostasis undergo continuous crosstalk. Complement, a component of innate immunity, is one such system. Initially regarded as a system to protect the host from infection, complement has more recently been shown to have numerous other functions, including involvement in embryonic development, tissue modeling, and repair. Furthermore, the complement system plays a major role in the pathophysiology of many diseases. Through interactions with other plasma cascades, including hemostasis, complement activation leads to the broad host-protective response known as thromboinflammation. Most complement research has been limited to reductionistic models of purified components and cells and their interactions in vitro. However, to study the pathophysiology of complement-driven diseases, including the interaction between the complement system and other inflammatory systems, holistic models demonstrating only minimal interference with complement activity are needed. Here we describe two such models; whole blood anticoagulated with either the thrombin inhibitor lepirudin or the fibrin polymerization peptide blocker GPRP, both of which retain complement activity and preserve the ability of complement to be mutually reactive with other inflammatory systems. For instance, to examine the relative roles of C3 and C5 in complement activation, it is possible to compare the effects of the C3 inhibitor compstatin effects to those of inhibitors of C5 and C5aR1. We also discuss how complement is activated by both pathogen-associated molecular patterns, inducing infectious inflammation caused by organisms such as Gram-negative and Gram-positive bacteria, and by sterile damage-associated molecular patterns, including cholesterol crystals and artificial materials used in clinical medicine. When C3 is inhibited, it is important to determine the mechanism by which inflammation is attenuated, i.e., whether the attenuation derives directly from C3 activation products or via downstream activation of C5, since the mechanism involved may determine the appropriate choice of inhibitor under various conditions. With some exceptions, most inflammatory responses are dependent on C5 and C5aR1; one exception is venous air embolism, in which air bubbles enter the blood circulation and trigger a mainly C3-dependent thromboembolism, with the formation of an active C3 convertase, without a corresponding C5 activation. Under such conditions, an inhibitor of C3 is needed to attenuate the inflammation. Our holistic blood models will be useful for further studies of the inhibition of any complement target, not just C3 or C5. The focus here will be on targeting the critical complement component, activation product, or receptor that is important for the pathophysiology in a variety of disease conditions.
Collapse
Affiliation(s)
- Tom E Mollnes
- Research Laboratory, Nordland Hospital, Bodø, Norway; Department of Immunology, Oslo University Hospital and University of Oslo, Norway; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Benjamin S Storm
- Research Laboratory, Nordland Hospital, Bodø, Norway; Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway; Faculty of Nursing and Health Sciences, Nord University, Bodø, Norway
| | - Ole L Brekke
- Research Laboratory, Nordland Hospital, Bodø, Norway; Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway; Department of Laboratory Medicine, Nordland Hospital, Bodø, Norway
| | - Per H Nilsson
- Department of Immunology, Oslo University Hospital and University of Oslo, Norway; Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, 39182 Kalmar, Sweden; Department of Chemistry and Biomedical Sciences, Linnaeus University, 39182 Kalmar, Sweden
| | - John D Lambris
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
21
|
Mossanen Parsi M, Duval C, Ariëns RAS. Vascular Dementia and Crosstalk Between the Complement and Coagulation Systems. Front Cardiovasc Med 2021; 8:803169. [PMID: 35004913 PMCID: PMC8733168 DOI: 10.3389/fcvm.2021.803169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 01/12/2023] Open
Abstract
Vascular Dementia (VaD) is a neurocognitive disorder caused by reduced blood flow to the brain tissue, resulting in infarction, and is the second most common type of dementia. The complement and coagulation systems are evolutionary host defence mechanisms activated by acute tissue injury to induce inflammation, clot formation and lysis; recent studies have revealed that these systems are closely interlinked. Overactivation of these systems has been recognised to play a key role in the pathogenesis of neurological disorders such as Alzheimer's disease and multiple sclerosis, however their role in VaD has not yet been extensively reviewed. This review aims to bridge the gap in knowledge by collating current understanding of VaD to enable identification of complement and coagulation components involved in the pathogenesis of this disorder that may have their effects amplified or supressed by crosstalk. Exploration of these mechanisms may unveil novel therapeutic targets or biomarkers that would improve current treatment strategies for VaD.
Collapse
Affiliation(s)
| | | | - Robert A. S. Ariëns
- Discovery and Translational Science Department, School of Medicine, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
22
|
Barale C, Melchionda E, Morotti A, Russo I. Prothrombotic Phenotype in COVID-19: Focus on Platelets. Int J Mol Sci 2021; 22:ijms222413638. [PMID: 34948438 PMCID: PMC8705811 DOI: 10.3390/ijms222413638] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 infection is associated with a broad spectrum of presentations, but alveolar capillary microthrombi have been described as a common finding in COVID-19 patients, appearing as a consequence of a severe endothelial injury with endothelial cell membrane disruption. These observations clearly point to the identification of a COVID-19-associated coagulopathy, which may contribute to thrombosis, multi-organ damage, and cause of severity and fatality. One significant finding that emerges in prothrombotic abnormalities observed in COVID-19 patients is that the coagulation alterations are mainly mediated by the activation of platelets and intrinsically related to viral-mediated endothelial inflammation. Beyond the well-known role in hemostasis, the ability of platelets to also release various potent cytokines and chemokines has elevated these small cells from simple cell fragments to crucial modulators in the blood, including their inflammatory functions, that have a large influence on the immune response during infectious disease. Indeed, platelets are involved in the pathogenesis of acute lung injury also by promoting NET formation and affecting vascular permeability. Specifically, the deposition by activated platelets of the chemokine platelet factor 4 at sites of inflammation promotes adhesion of neutrophils on endothelial cells and thrombogenesis, and it seems deeply involved in the phenomenon of vaccine-induced thrombocytopenia and thrombosis. Importantly, the hyperactivated platelet phenotype along with evidence of cytokine storm, high levels of P-selectin, D-dimer, and, on the other hand, decreased levels of fibrinogen, von Willebrand factor, and thrombocytopenia may be considered suitable biomarkers that distinguish the late stage of COVID-19 progression in critically ill patients.
Collapse
Affiliation(s)
| | | | | | - Isabella Russo
- Correspondence: ; Tel.: +39-011-6705447; Fax: +39-011-9038639
| |
Collapse
|
23
|
Berkowitz S, Chapman J, Dori A, Gofrit SG, Maggio N, Shavit-Stein E. Complement and Coagulation System Crosstalk in Synaptic and Neural Conduction in the Central and Peripheral Nervous Systems. Biomedicines 2021; 9:biomedicines9121950. [PMID: 34944766 PMCID: PMC8698364 DOI: 10.3390/biomedicines9121950] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
Complement and coagulation are both key systems that defend the body from harm. They share multiple features and are similarly activated. They each play individual roles in the systemic circulation in physiology and pathophysiology, with significant crosstalk between them. Components from both systems are mapped to important structures in the central nervous system (CNS) and peripheral nervous system (PNS). Complement and coagulation participate in critical functions in neuronal development and synaptic plasticity. During pathophysiological states, complement and coagulation factors are upregulated and can modulate synaptic transmission and neuronal conduction. This review summarizes the current evidence regarding the roles of the complement system and the coagulation cascade in the CNS and PNS. Possible crosstalk between the two systems regarding neuroinflammatory-related effects on synaptic transmission and neuronal conduction is explored. Novel treatment based on the modulation of crosstalk between complement and coagulation may perhaps help to alleviate neuroinflammatory effects in diseased states of the CNS and PNS.
Collapse
Affiliation(s)
- Shani Berkowitz
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Joab Chapman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Robert and Martha Harden Chair in Mental and Neurological Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Amir Dori
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan 6997801, Israel
| | - Shany Guly Gofrit
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Correspondence: ; Tel.: +972-50-921-0400
| |
Collapse
|
24
|
Pitkänen HH, Jouppila A, Helin T, Dulipati V, Kotimaa J, Meri S, Kantele A, Jalkanen P, Julkunen I, Lassila R. COVID-19 adenovirus vaccine triggers antibodies against PF4 complexes to activate complement and platelets. Thromb Res 2021; 208:129-137. [PMID: 34768097 PMCID: PMC8571998 DOI: 10.1016/j.thromres.2021.10.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/14/2021] [Accepted: 10/28/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Vaccine-induced thrombotic thrombocytopenia (VITT) is a rare coagulation disorder reported after administration of COVID-19 adenovirus-vectored vaccines. VITT is mediated by anti-platelet factor 4 (PF4) antibodies activating platelets through the Fcγ-receptor II (FcγRII), and it is associated with strong fibrin turnover. The complement system is involved in several other immunothrombotic entities, but its impact on VITT is not established. OBJECTIVE To assess antibodies in interaction with the activation of platelets and complement triggered by VITT. METHODS Antibodies against adenovirus type 2 hexon protein, ChAdOx1 adenoviral vector-specific IgG and PF4 were analyzed by enzyme immunoassays from VITT patients (n = 5). The EDTA plasma samples of the patients and controls were used to measure both terminal complement complexes (TCC) by ELISA and aggregation of healthy donor platelets. We studied the effects of human immunoglobulin (IVIG) and glycoprotein IIb/IIIa inhibitor (GPIIb/IIIa) on spontaneous and collagen-induced platelet aggregation supplemented with VITT plasma. RESULTS None of the patients had experienced a COVID-19 infection. Antibody analyses confirmed the immunogenicity of the adenovirus-vectored ChAdOx1 vaccine. Moreover, VITT plasma had anti-PF4 antibodies and elevated TCC levels as a sign of complement activation. In isolated healthy donor platelets, VITT patient plasma caused marked, spontaneous aggregation of platelets, which was abolished by eptifibatide and high-dose therapeutic IVIG. CONCLUSIONS Our findings suggest that VITT is triggered by antibodies against adenovirus vector and PF4-polyanion complexes which strongly co-activate complement and platelets. The spontaneous platelet aggregation was suppressed by IVIG or eptifibatide, indicating that besides FcγRII, also GPIIb/IIIa receptor exerts platelet procoagulant role in VITT.
Collapse
Affiliation(s)
- Hanna H. Pitkänen
- Helsinki University, Division of Anesthesiology, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland,Clinical Research Institute Helsinki University Hospital, Helsinki, Finland
| | - Annukka Jouppila
- Clinical Research Institute Helsinki University Hospital, Helsinki, Finland
| | - Tuukka Helin
- HUSLAB, Clinical Chemistry, Helsinki University Hospital and University of Helsinki
| | - Vinaya Dulipati
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Finland
| | - Juha Kotimaa
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Finland
| | - Seppo Meri
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Finland,HUSLAB, Helsinki University Hospital, Finland
| | - Anu Kantele
- Meilahti Infectious Diseases and Vaccine Research Center, MeVac, Department of Infectious Diseases, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Pinja Jalkanen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine, University of Turku, Turku, Finland,Turku University Hospital, Clinical Microbiology, Turku, Finland
| | - Riitta Lassila
- Department of Hematology, Coagulation Disorders Unit, Helsinki University Hospital, Helsinki, Finland,Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Finland,Finnish Institute for Health and Welfare, Helsinki, Finland,Corresponding author at: Coagulation Disorders Unit, Department of Hematology and Comprehensive Cancer Center, Laboratory Services, HUCH, PL 372, 00029 Helsinki, Finland
| |
Collapse
|
25
|
Lim MS, Mcrae S. COVID-19 and immunothrombosis: Pathophysiology and therapeutic implications. Crit Rev Oncol Hematol 2021; 168:103529. [PMID: 34800652 PMCID: PMC8596655 DOI: 10.1016/j.critrevonc.2021.103529] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 09/11/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022] Open
Abstract
The coagulopathy of COVID-19 is characterised by significantly elevated D Dimer and fibrinogen, mild thrombocytopenia and a mildly prolonged PT/APTT. A high incidence of thrombotic complications occurs despite standard thromboprophylaxis. The evidence to date supports immunothrombosis as the underlying mechanism for this coagulopathy which is triggered by a hyperinflammatory response and endotheliopathy. A hypercoagulable state results from endothelial damage/activation, complement activation, platelet hyperactivity, release of Extracellular Neutrophil Traps, activation of the coagulation system and a "hypofibrinolytic" state. Significant cross-talk occurs between the innate/adaptive immune system, endothelium and the coagulation system. D dimer has been shown to be the most reliable predictor of disease severity, thrombosis, and overall survival. In this context, targeting pathways upstream of coagulation using novel or repurposed drugs alone or in combination with other anti-thrombotic agents may be a rational approach to prevent the mortality/morbidity due to COVID-19 associated coagulopathy.
Collapse
Affiliation(s)
- Ming Sheng Lim
- Department of Hematology, Launceston General Hospital, WP Holman Clinic, Level 1. PO Box 1963, Launceston, Tasmania, Australia.
| | - Simon Mcrae
- Department of Hematology, Launceston General Hospital, WP Holman Clinic, Level 1. PO Box 1963, Launceston, Tasmania, Australia.
| |
Collapse
|
26
|
Steadman E, Fandaros M, Yin W. SARS-CoV-2 and Plasma Hypercoagulability. Cell Mol Bioeng 2021; 14:513-522. [PMID: 34221178 PMCID: PMC8238024 DOI: 10.1007/s12195-021-00685-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Hypercoagulability has emerged as a prominent consequence of COVID-19. This presents challenges not only in the clinic, but also in thrombosis research. Health and safety considerations, the status of the blood and plasma supply, the infection status of individual donors, and the mechanisms by which SARS-CoV-2 activates coagulation are all of concern. In this review, we discuss these topics from the basic research perspective. As in other respiratory illnesses, blood and plasma from COVID-19 positive patients carries minimal to no risk of infection to practitioners or researchers. There are currently no special regulatory mandates directing individual donors (for research purposes), blood centers/services or vendors (for blood products for research) to test blood/plasma for SARS-CoV-2 or antibodies. We discuss current theories about how SARS-CoV-2 leads to hyper-coagulant state in severe cases of COVID-19. Our current understanding of the mechanisms behind COVID-19 associated thromboembolic events have centered around three different pathways: (1) direct activation of platelets, enhancing coagulation; (2) direct infection and indirect activation (e.g. cytokine storm) of endothelial cells by SARS-CoV-2, shifting endothelium from an anti-thrombotic to a pro-thrombotic state; and (3) direct activation of complement pathways, promoting thrombin generation. Further investigation on how SARS-CoV-2 affects thrombosis in COVID-19 patients may bring novel anti-thrombotic therapies to combat the disease.
Collapse
Affiliation(s)
- Elisabeth Steadman
- Department of Biomedical Engineering, Stony Brook University, Bioengineering Building, Room 109, Stony Brook, NY 11794 USA
| | - Marina Fandaros
- Department of Biomedical Engineering, Stony Brook University, Bioengineering Building, Room 109, Stony Brook, NY 11794 USA
| | - Wei Yin
- Department of Biomedical Engineering, Stony Brook University, Bioengineering Building, Room 109, Stony Brook, NY 11794 USA
| |
Collapse
|
27
|
Fandaros M, Joseph K, Kaplan AP, Rubenstein DA, Ghebrehiwet B, Yin W. gC1qR Antibody Can Modulate Endothelial Cell Permeability in Angioedema. Inflammation 2021; 45:116-128. [PMID: 34494203 DOI: 10.1007/s10753-021-01532-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/23/2021] [Accepted: 07/31/2021] [Indexed: 10/20/2022]
Abstract
Angioedema is characterized by swelling of the skin or mucous membranes. Overproduction of the vasodilator bradykinin (BK) is an important contributor to the disease pathology, which causes rapid increase in vascular permeability. BK formation on endothelial cells results from high molecular weight kininogen (HK) interacting with gC1qR, the receptor for the globular heads of C1q, the first component of the classical pathway of complement. Endothelial cells are sensitive to blood-flow-induced shear stress and it has been shown that shear stress can modulate gC1qR expression. This study aimed to determine the following: (1) how BK or angioedema patients' (HAE) plasma affected endothelial cell permeability and gC1qR expression under shear stress, and (2) if monoclonal antibody (mAb) 74.5.2, which recognizes the HK binding site on gC1qR, had an inhibitory effect in HK binding to endothelial cells. Human dermal microvascular endothelial cells (HDMECs) grown on Transwell inserts were exposed to shear stress in the presence of HAE patients' plasma. Endothelial cell permeability was measured using FITC-conjugated bovine serum albumin. gC1qR expression and HK binding to endothelial cell surface was measured using solid-phase ELISA. Cell morphology was quantified using immunofluorescence microscopy. The results demonstrated that BK at 1 µg/mL, but not HAE patients' plasma and/or shear stress, caused significant increases in HDMEC permeability. The mAb 74.5.2 could effectively inhibit HK binding to recombinant gC1qR, and reduce HAE patients' plasma-induced HDMEC permeability change. These results suggested that monoclonal antibody to gC1qR, i.e., 74.5.2, could be potentially used as an effective therapeutic reagent to prevent angioedema.
Collapse
Affiliation(s)
- Marina Fandaros
- Department of Biomedical Engineering, Stony Brook University, NY, Stony Brook, USA
| | - Kusumam Joseph
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA.,BioCryst Pharmaceuticals Inc., Durham, NC, 27703, USA
| | - Allen P Kaplan
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - David A Rubenstein
- Department of Biomedical Engineering, Stony Brook University, NY, Stony Brook, USA
| | | | - Wei Yin
- Department of Biomedical Engineering, Stony Brook University, NY, Stony Brook, USA.
| |
Collapse
|
28
|
Sommerfeld O, Dahlke K, Sossdorf M, Claus RA, Scherag A, Bauer M, Bloos F. Complement factor D is linked to platelet activation in human and rodent sepsis. Intensive Care Med Exp 2021; 9:41. [PMID: 34396466 PMCID: PMC8364893 DOI: 10.1186/s40635-021-00405-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The complement factor D (CFD) exerts a regulatory role during infection. However, its physiological function in coagulopathy and its impact on the course of an infection remains unclear. MATERIALS Wild-type and CFD-deficient mice (n = 91) were subjected to cecal ligation and puncture to induce sepsis. At several time points, markers of coagulation and the host-immune response were determined. Furthermore, in patients (n = 79) with sepsis or SIRS, CFD levels were related to clinical characteristics, use of antiplatelet drugs and outcome. RESULTS Septic CFD-deficient mice displayed higher TAT complexes (p = 0.02), impaired maximal clot firmness, but no relevant platelet drop and reduced GPIIb/IIIa surface expression on platelets (p = 0.03) compared to septic wild-type mice. In humans, higher CFD levels (non-survivors, 5.0 µg/ml to survivors, 3.6 µg/ml; p = 0.015) were associated with organ failure (SOFA score: r = 0.33; p = 0.003) and mortality (75% percentile, 61.1% to 25% percentile, 26.3%). CFD level was lower in patients with antiplatelet drugs (4.5-5.3 µg/ml) than in patients without. CONCLUSION In mice, CFD is linked to pronounced platelet activation, depicted by higher GPIIb/IIIa surface expression in wild-type mice. This might be of clinical importance since high CFD plasma concentrations were also associated with increased mortality in sepsis patients.
Collapse
Affiliation(s)
- O Sommerfeld
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany. .,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.
| | - K Dahlke
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - M Sossdorf
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - R A Claus
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - A Scherag
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Institute of Medical Statistics, Computer and Data Sciences, Jena University Hospital, Jena, Germany
| | - M Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - F Bloos
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany. .,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.
| |
Collapse
|
29
|
van Zanden JE, Jager NM, Seelen MA, Daha MR, Veldhuis ZJ, Leuvenink HG, Erasmus ME. Brain death-induced lung injury is complement dependent, with a primary role for the classical/lectin pathway. Am J Transplant 2021; 21:993-1002. [PMID: 32743873 PMCID: PMC7984080 DOI: 10.1111/ajt.16231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 01/25/2023]
Abstract
In brain-dead donors immunological activation occurs, which deteriorates donor lung quality. Whether the complement system is activated and which pathways are herein involved, remain unknown. We aimed to investigate whether brain death (BD)-induced lung injury is complement dependent and dissected the contribution of the complement activation pathways. BD was induced and sustained for 3 hours in wild-type (WT) and complement deficient mice. C3-/- mice represented total complement deficiency, C4-/- mice represented deficiency of the classical and lectin pathway, and factor properdin (P)-/- mice represented alternative pathway deficiency. Systemic and local complement levels, histological lung injury, and pulmonary inflammation were assessed. Systemic and local complement levels were reduced in C3-/- mice. In addition, histological lung injury and inflammation were attenuated, as corroborated by influx of neutrophils and gene expressions of interleukin (IL)-6, IL-8-like KC, TNF-α, E-selectin, and MCP-1. In C4-/- mice, complement was reduced on both systemic and local levels and histological lung injury and inflammatory status were ameliorated. In P-/- mice, histological lung injury was attenuated, though systemic and local complement levels, IL-6 and KC gene expressions, and neutrophil influx were not affected. We demonstrated that BD-induced lung injury is complement dependent, with a primary role for the classical/lectin activation pathway.
Collapse
Affiliation(s)
- Judith E. van Zanden
- Department of SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Neeltina M. Jager
- Department of SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Marc A. Seelen
- Division of NephrologyDepartment of Internal MedicineUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Mohamed R. Daha
- Division of NephrologyDepartment of Internal MedicineUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands,Department of NephrologyLeiden University Medical CenterLeidenthe Netherlands
| | - Zwanida J. Veldhuis
- Department of SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Henri G.D. Leuvenink
- Department of SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Michiel E. Erasmus
- Department of Cardiothoracic SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| |
Collapse
|
30
|
Magro CM, Mulvey J, Kubiak J, Mikhail S, Suster D, Crowson AN, Laurence J, Nuovo G. Severe COVID-19: A multifaceted viral vasculopathy syndrome. Ann Diagn Pathol 2021; 50:151645. [PMID: 33248385 PMCID: PMC7553104 DOI: 10.1016/j.anndiagpath.2020.151645] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/09/2020] [Indexed: 01/08/2023]
Abstract
The objective of this study was to elucidate the pathophysiology that underlies severe COVID-19 by assessing the histopathology and the in situ detection of infectious SARS-CoV-2 and viral capsid proteins along with the cellular target(s) and host response from twelve autopsies. There were three key findings: 1) high copy infectious virus was limited mostly to the alveolar macrophages and endothelial cells of the septal capillaries; 2) viral spike protein without viral RNA localized to ACE2+ endothelial cells in microvessels that were most abundant in the subcutaneous fat and brain; 3) although both infectious virus and docked viral spike protein was associated with complement activation, only the endocytosed pseudovirions induced a marked up-regulation of the key COVID-19 associated proteins IL6, TNF alpha, IL1 beta, p38, IL8, and caspase 3. Importantly, this microvasculitis was associated with characteristic findings on hematoxylin and eosin examination that included endothelial degeneration and resultant basement membrane zone disruption and reduplication. It is concluded that serious COVID-19 infection has two distinct mechanisms: 1) a microangiopathy of pulmonary capillaries associated with a high infectious viral load where endothelial cell death releases pseudovirions into the circulation, and 2) the pseudovirions dock on ACE2+ endothelial cells most prevalent in the skin/subcutaneous fat and brain that activates the complement pathway/coagulation cascade resulting in a systemic procoagulant state as well as the expression of cytokines that produce the cytokine storm. The data predicts a favorable response to therapies based on either removal of circulating viral proteins and/or blunting of the endothelial-induced response.
Collapse
Affiliation(s)
- Cynthia M Magro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NY, NY, USA
| | - Justin Mulvey
- Department of Laboratory Medicine, Memorial Sloan-Kettering Cancer Center, NY, NY, USA
| | - Jeffrey Kubiak
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NY, NY, USA
| | | | - David Suster
- Rutgers University Hospital Department of Pathology, Newark, NJ, USA
| | - A Neil Crowson
- Pathology Laboratory Associates, Oklahoma City, OK, USA; University of Oklahoma, Oklahoma City, OK, USA
| | - Jeffrey Laurence
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NY, NY, USA
| | - Gerard Nuovo
- Discovery Life Sciences, Powell, OH, USA; Ohio State University Comprehensive Cancer Center and Discovery Life Sciences, Columbus, OH, USA.
| |
Collapse
|
31
|
Maxwell AJ, Ding J, You Y, Dong Z, Chehade H, Alvero A, Mor Y, Draghici S, Mor G. Identification of key signaling pathways induced by SARS-CoV2 that underlie thrombosis and vascular injury in COVID-19 patients. J Leukoc Biol 2021; 109:35-47. [PMID: 33242368 PMCID: PMC7753679 DOI: 10.1002/jlb.4covr0920-552rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/05/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022] Open
Abstract
The SARS-CoV-2 pandemic has led to hundreds of thousands of deaths and billions of dollars in economic damage. The immune response elicited from this virus is poorly understood. An alarming number of cases have arisen where COVID-19 patients develop complications on top of the symptoms already associated with SARS, such as thrombosis, injuries of vascular system, kidney, and liver, as well as Kawasaki disease. In this review, a bioinformatics approach was used to elucidate the immune response triggered by SARS-CoV-2 infection in primary human lung epithelial and transformed human lung alveolar. Additionally, examined the potential mechanism behind several complications that have been associated with COVID-19 and determined that a specific cytokine storm is leading to excessive neutrophil recruitment. These neutrophils are directly leading to thrombosis, organ damage, and complement activation via neutrophil extracellular trap release.
Collapse
Affiliation(s)
- Anthony J Maxwell
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Jiahui Ding
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Yuan You
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Zhong Dong
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Ayesha Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Yechiel Mor
- Department of Internal Medicine Wayne State University, Detroit, Michigan, USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, Michigan, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
32
|
Gautam I, Storad Z, Filipiak L, Huss C, Meikle CK, Worth RG, Wuescher LM. From Classical to Unconventional: The Immune Receptors Facilitating Platelet Responses to Infection and Inflammation. BIOLOGY 2020; 9:E343. [PMID: 33092021 PMCID: PMC7589078 DOI: 10.3390/biology9100343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/06/2020] [Accepted: 10/15/2020] [Indexed: 12/14/2022]
Abstract
Platelets have long been recognized for their role in maintaining the balance between hemostasis and thrombosis. While their contributions to blood clotting have been well established, it has been increasingly evident that their roles extend to both innate and adaptive immune functions during infection and inflammation. In this comprehensive review, we describe the various ways in which platelets interact with different microbes and elicit immune responses either directly, or through modulation of leukocyte behaviors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Leah M. Wuescher
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (I.G.); (Z.S.); (L.F.); (C.H.); (C.K.M.); (R.G.W.)
| |
Collapse
|
33
|
Jo WY, Oh H, Kim H, Yoon HK, Lee HC, Cho WS, Kim JE, Park HP. Preoperative platelet count may predict postoperative symptomatic cerebral hyperperfusion syndrome after superficial temporal artery-middle cerebral artery anastomosis in moyamoya patients. Clin Neurol Neurosurg 2020; 199:106260. [PMID: 33031987 DOI: 10.1016/j.clineuro.2020.106260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/21/2020] [Accepted: 09/29/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Platelets play a critical role in the inflammatory response, accompanied by microvascular endothelial dysfunction, underlying postoperative symptomatic cerebral hyperperfusion syndrome (PSCHS) after superficial temporal artery-middle cerebral artery (STA-MCA) anastomosis in moyamoya patients. We examined whether the preoperative platelet count can predict PSCHS after STA-MCA anastomosis in such patients. METHODS In 160 adult moyamoya patients undergoing 186 STA-MCA anastomoses, preoperative (demographics, initial clinical manifestation, and Suzuki grade), intraoperative (surgical time, operative side, fluid balance, and maximum and minimum mean blood pressure before and after vessel anastomosis), immediate postoperative (APACHE 2 score), and laboratory (hemoglobin and C-reactive protein levels and white blood cell and platelet counts) data were collected retrospectively. RESULTS 84 patients (90 sides, 48.4 %) developed PSCHS with a median(IQR) onset of postoperative day 1(0-3) and duration of 4(3-7) days. The preoperative (25.2[22.8-28.0] vs. 23.1[19.7-26.2] ×104/μL, p = 0.009) platelet count was significantly higher in patients with PSCHS than in those without. The preoperative platelet count (odds ratio[95 % confidence interval], 1.14[1.03-1.27], p = 0.011), operation on the dominant hemisphere (6.84 [3.26-14.36], p < 0.001), and negative fluid balance (2.41[1.04-5.59], p = 0.040) were significant independent predictors of PSCHS. The optimal cut-off value for preoperative platelet count was 22.7 ×104/μL, and PSCHS developed more frequently in cases with a preoperative platelet count ≥ 22.7 × 104/μL (2.90[1.54-5.45]; p = 0.001). CONCLUSION A high preoperative platelet count may be associated with the development of PSCHS after STA-MCA anastomosis in adult moyamoya patients.
Collapse
Affiliation(s)
- Woo-Young Jo
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyongmin Oh
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Heewon Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyun-Kyu Yoon
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyung-Chul Lee
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Won-Sang Cho
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Jeong-Eun Kim
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Hee-Pyoung Park
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
34
|
Does Complement-Mediated Hemostatic Disturbance Occur in Traumatic Brain Injury? A Literature Review and Observational Study Protocol. Int J Mol Sci 2020; 21:ijms21051596. [PMID: 32111078 PMCID: PMC7084711 DOI: 10.3390/ijms21051596] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/07/2020] [Accepted: 02/25/2020] [Indexed: 12/11/2022] Open
Abstract
Despite improvements in medical triage and tertiary care, traumatic brain injury (TBI) remains associated with significant morbidity and mortality. Almost two-thirds of patients with severe TBI develop some form of hemostatic disturbance, which contributes to poor outcome. In addition, the complement system, which is abundant in the healthy brain, undergoes significant intra- and extracranial amplification following TBI. Previously considered to be structurally similar but separate systems, evidence of an interaction between the complement and coagulation systems in non-TBI cohorts has accumulated, with the activation of one system amplifying the activation of the other, independent of their established pathways. However, it is not known whether this interaction exists in TBI. In this review we summarize the available literature on complement activation following TBI, and the crosstalk between the complement and coagulation systems. We demonstrate how the complement system interacts with the coagulation cascade by activating the intrinsic coagulation pathway and by bypassing the initial cascade and directly producing thrombin as well. This crosstalk also effects platelets, where evidence points to a relationship with the complement system on multiple levels, with complement anaphylatoxins being able to induce disproportionate platelet activation and adhesion. The complement system also stimulates thrombosis by inhibiting fibrinolysis and stimulating endothelial cells to release prothrombotic microparticles. These interactions see clinical relevance in several disorders where a deficiency in complement regulation seems to result in a prothrombotic clinical presentation. Finally, based on these observations, we present the outline of an observational cohort study that is currently under preparation and aimed at assessing how complement influences coagulation in patients with isolated TBI.
Collapse
|
35
|
Ramadori P, Klag T, Malek NP, Heikenwalder M. Platelets in chronic liver disease, from bench to bedside. JHEP Rep 2019; 1:448-459. [PMID: 32039397 PMCID: PMC7005648 DOI: 10.1016/j.jhepr.2019.10.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/04/2019] [Accepted: 10/09/2019] [Indexed: 02/07/2023] Open
Abstract
In the last decade, numerous studies revealed physiologic and pathophysiologic roles of platelets beyond haemostasis, a process to prevent and stop bleeding. These include the activation of the immune system and the promotion of inflammation, infection and cancer. Hence, the emerging view on the role of platelets has shifted - platelets are now seen as alert "sentinels" of the immune compartment, rather than passive bystanders. Herein, we review well-established and newly discovered features of platelets that define their natural role in maintaining blood haemostasis, but also their functional relationship with other cells of the immune system. We focus on recent studies underlining functional involvement of platelets in chronic liver diseases and cancer, as well as the effects of anti-platelet therapy in these contexts. Finally, we illustrate the potential of platelets as possible diagnostic and therapeutic tools in liver disease based on recently developed methodologies.
Collapse
Affiliation(s)
- Pierluigi Ramadori
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | - Thomas Klag
- Department of Internal Medicine I, University of Tuebingen, Tuebingen, Germany
| | - Nisar Peter Malek
- Department of Internal Medicine I, University of Tuebingen, Tuebingen, Germany
- Corresponding authors. Address: Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany, Tel.: 0049-6221423891, or Department of Internal Medicine I, University Hospital of Tuebingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany, Tel.: 0049-70712982721.
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
- Corresponding authors. Address: Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany, Tel.: 0049-6221423891, or Department of Internal Medicine I, University Hospital of Tuebingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany, Tel.: 0049-70712982721.
| |
Collapse
|
36
|
Sauter RJ, Sauter M, Reis ES, Emschermann FN, Nording H, Ebenhöch S, Kraft P, Münzer P, Mauler M, Rheinlaender J, Madlung J, Edlich F, Schäffer TE, Meuth SG, Duerschmied D, Geisler T, Borst O, Gawaz M, Kleinschnitz C, Lambris JD, Langer HF. Functional Relevance of the Anaphylatoxin Receptor C3aR for Platelet Function and Arterial Thrombus Formation Marks an Intersection Point Between Innate Immunity and Thrombosis. Circulation 2019; 138:1720-1735. [PMID: 29802205 DOI: 10.1161/circulationaha.118.034600] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Platelets have distinct roles in the vascular system in that they are the major mediator of thrombosis, critical for restoration of tissue integrity, and players in vascular inflammatory conditions. In close spatiotemporal proximity, the complement system acts as the first line of defense against invading microorganisms and is a key mediator of inflammation. Whereas the fluid phase cross-talk between the complement and coagulation systems is well appreciated, the understanding of the pathophysiological implications of such interactions is still scant. METHODS We analyzed coexpression of the anaphylatoxin receptor C3aR with activated glycoprotein IIb/IIIa on platelets of 501 patients with coronary artery disease using flow cytometry; detected C3aR expression in human or murine specimen by polymerase chain reaction, immunofluorescence, Western blotting, or flow cytometry; and examined the importance of platelet C3aR by various in vitro platelet function tests, in vivo bleeding time, and intravital microscopy. The pathophysiological relevance of C3aR was scrutinized with the use of disease models of myocardial infarction and stroke. To approach underlying molecular mechanisms, we identified the platelet small GTPase Rap1b using nanoscale liquid chromatography coupled to tandem mass spectrometry. RESULTS We found a strong positive correlation of platelet complement C3aR expression with activated glycoprotein IIb/IIIa in patients with coronary artery disease and coexpression of C3aR with glycoprotein IIb/IIIa in thrombi obtained from patients with myocardial infarction. Our results demonstrate that the C3a/C3aR axis on platelets regulates distinct steps of thrombus formation such as platelet adhesion, spreading, and Ca2+ influx. Using C3aR-/- mice or C3-/- mice with reinjection of C3a, we uncovered that the complement activation fragment C3a regulates bleeding time after tail injury and thrombosis. Notably, C3aR-/- mice were less prone to experimental stroke and myocardial infarction. Furthermore, reconstitution of C3aR-/- mice with C3aR+/+ platelets and platelet depletion experiments demonstrated that the observed effects on thrombosis, myocardial infarction, and stroke were specifically caused by platelet C3aR. Mechanistically, C3aR-mediated signaling regulates the activation of Rap1b and thereby bleeding arrest after injury and in vivo thrombus formation. CONCLUSIONS Overall, our findings uncover a novel function of the anaphylatoxin C3a for platelet function and thrombus formation, highlighting a detrimental role of imbalanced complement activation in cardiovascular diseases.
Collapse
Affiliation(s)
- Reinhard J Sauter
- Department of Cardiology and Cardiovascular Medicine, University Clinic (R.J.S., H.N., P.M., T.G., O.B., M.G., H.F.L.), Eberhard Karls-University Tübingen, Germany.,Section for Cardioimmunology (R.J.S., M.S., F.N.E., H.N., S.E., H.F.L.), Eberhard Karls-University Tübingen, Germany
| | - Manuela Sauter
- Section for Cardioimmunology (R.J.S., M.S., F.N.E., H.N., S.E., H.F.L.), Eberhard Karls-University Tübingen, Germany
| | - Edimara S Reis
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania, Philadelphia (E.S.R., J.D.L.)
| | - Frederic N Emschermann
- Section for Cardioimmunology (R.J.S., M.S., F.N.E., H.N., S.E., H.F.L.), Eberhard Karls-University Tübingen, Germany
| | - Henry Nording
- Department of Cardiology and Cardiovascular Medicine, University Clinic (R.J.S., H.N., P.M., T.G., O.B., M.G., H.F.L.), Eberhard Karls-University Tübingen, Germany.,Section for Cardioimmunology (R.J.S., M.S., F.N.E., H.N., S.E., H.F.L.), Eberhard Karls-University Tübingen, Germany
| | - Sonja Ebenhöch
- Section for Cardioimmunology (R.J.S., M.S., F.N.E., H.N., S.E., H.F.L.), Eberhard Karls-University Tübingen, Germany
| | - Peter Kraft
- Department of Neurology, University of Würzburg, Germany (P.K.)
| | - Patrick Münzer
- Department of Cardiology and Cardiovascular Medicine, University Clinic (R.J.S., H.N., P.M., T.G., O.B., M.G., H.F.L.), Eberhard Karls-University Tübingen, Germany
| | - Maximilian Mauler
- Cardiology and Angiology I, Heart Center Freiburg University and Faculty of Medicine (M.M., D.D.), University of Freiburg, Germany
| | - Johannes Rheinlaender
- Institute of Applied Physics (J.R., T.E.S.), Eberhard Karls-University Tübingen, Germany
| | - Johannes Madlung
- Proteom Center, Interfaculty Institute for Cell Biology (J.M.), Eberhard Karls-University Tübingen, Germany
| | - Frank Edlich
- Institute of Biochemistry (F.E.), University of Freiburg, Germany.,Institute for Biochemistry and Molecular Biology, University of Freiburg, Germany (F.E.).,BIOSS, Centre for Biological Signaling Studies, University of Freiburg, Germany (F.E.)
| | - Tilman E Schäffer
- Institute of Applied Physics (J.R., T.E.S.), Eberhard Karls-University Tübingen, Germany
| | - Sven G Meuth
- Department of Neurology, University of Münster, Germany (S.G.M.)
| | - Daniel Duerschmied
- Cardiology and Angiology I, Heart Center Freiburg University and Faculty of Medicine (M.M., D.D.), University of Freiburg, Germany
| | - Tobias Geisler
- Department of Cardiology and Cardiovascular Medicine, University Clinic (R.J.S., H.N., P.M., T.G., O.B., M.G., H.F.L.), Eberhard Karls-University Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Cardiovascular Medicine, University Clinic (R.J.S., H.N., P.M., T.G., O.B., M.G., H.F.L.), Eberhard Karls-University Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Cardiovascular Medicine, University Clinic (R.J.S., H.N., P.M., T.G., O.B., M.G., H.F.L.), Eberhard Karls-University Tübingen, Germany
| | | | - John D Lambris
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania, Philadelphia (E.S.R., J.D.L.)
| | - Harald F Langer
- Department of Cardiology and Cardiovascular Medicine, University Clinic (R.J.S., H.N., P.M., T.G., O.B., M.G., H.F.L.), Eberhard Karls-University Tübingen, Germany.,Section for Cardioimmunology (R.J.S., M.S., F.N.E., H.N., S.E., H.F.L.), Eberhard Karls-University Tübingen, Germany
| |
Collapse
|
37
|
Leiter O, Walker TL. Platelets: The missing link between the blood and brain? Prog Neurobiol 2019; 183:101695. [PMID: 31550515 DOI: 10.1016/j.pneurobio.2019.101695] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/19/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023]
Abstract
It is becoming increasingly clear that interactions between the peripheral immune system and the central nervous system are important in maintaining healthy brain function. Platelets are small blood cells traditionally known for their role in wound healing. However, platelets have recently been shown to exhibit many alternative functions. In this perspective, we summarize the repertoire of platelet functions, focusing on how these cells contribute to the maintenance of brain homeostasis and propose the mechanisms via which they could communicate with brain cells, including exosome and microparticle release and receptor interactions at local sites. In particular, we highlight the potential role that platelets play in maintaining brain plasticity via the modulation of new neuron generation from neural precursor cells, an interaction which could have important implications in the development of therapeutic interventions to promote cognitive function in aging and disease.
Collapse
Affiliation(s)
- Odette Leiter
- Queensland Brain Institute (QBI), The University of Queensland, Brisbane 4072, Australia.
| | - Tara L Walker
- Queensland Brain Institute (QBI), The University of Queensland, Brisbane 4072, Australia.
| |
Collapse
|
38
|
Guo L, Rondina MT. The Era of Thromboinflammation: Platelets Are Dynamic Sensors and Effector Cells During Infectious Diseases. Front Immunol 2019; 10:2204. [PMID: 31572400 PMCID: PMC6753373 DOI: 10.3389/fimmu.2019.02204] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/30/2019] [Indexed: 12/12/2022] Open
Abstract
Platelets are anucleate cells produced by megakaryocytes. In recent years, a robust body of literature supports the evolving role of platelets as key sentinel and effector cells in infectious diseases, especially critical in bridging hemostatic, inflammatory, and immune continuums. Upon intravascular pathogen invasion, platelets can directly sense viral, parasitic, and bacterial infections through pattern recognition receptors and integrin receptors or pathogen: immunoglobulin complexes through Fc and complement receptors—although our understanding of these interactions remains incomplete. Constantly scanning for areas of injury or inflammation as they circulate in the vasculature, platelets also indirectly respond to pathogen invasion through interactions with leukocytes and the endothelium. Following antigen recognition, platelets often become activated. Through a diverse repertoire of mechanisms, activated platelets can directly sequester or kill pathogens, or facilitate pathogen clearance by activating macrophages and neutrophils, promoting neutrophil extracellular traps (NETs) formation, forming platelet aggregates and microthrombi. At times, however, platelet activation may also be injurious to the host, exacerbating inflammation and promoting endothelial damage and thrombosis. There are many gaps in our understandings of the role of platelets in infectious diseases. However, with the emergence of advanced technologies, our knowledge is increasing. In the current review, we mainly discuss these evolving roles of platelets under four different infectious pathogen infections, of which are dengue, malaria, Esterichia coli (E. coli) and staphylococcus aureus S. aureus, highlighting the complex interplay of these processes with hemostatic and thrombotic pathways.
Collapse
Affiliation(s)
- Li Guo
- University of Utah Molecular Medicine Program, Salt Lake City, UT, United States
| | - Matthew T Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, UT, United States.,Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States.,Department of Pathology, University of Utah, Salt Lake City, UT, United States.,George E. Wahlen VAMC Department of Internal Medicine and GRECC, Salt Lake City, UT, United States
| |
Collapse
|
39
|
Abstract
Dysregulation of lymphocyte function, accumulation of autoantibodies and defective clearance of circulating immune complexes and apoptotic cells are hallmarks of systemic lupus erythematosus (SLE). Moreover, it is now evident that an intricate interplay between the adaptive and innate immune systems contributes to the pathogenesis of SLE, ultimately resulting in chronic inflammation and organ damage. Platelets circulate in the blood and are chiefly recognized for their role in the prevention of bleeding and promotion of haemostasis; however, accumulating evidence points to a role for platelets in both adaptive and innate immunity. Through a broad repertoire of receptors, platelets respond promptly to immune complexes, complement and damage-associated molecular patterns, and represent a major reservoir of immunomodulatory molecules in the circulation. Furthermore, evidence suggests that platelets are activated in patients with SLE, and that they could contribute to the circulatory autoantigenic load through the release of microparticles and mitochondrial antigens. Herein, we highlight how platelets contribute to the immune response and review evidence implicating platelets in the pathogenesis of SLE.
Collapse
|
40
|
Gorbet M, Sperling C, Maitz MF, Siedlecki CA, Werner C, Sefton MV. The blood compatibility challenge. Part 3: Material associated activation of blood cascades and cells. Acta Biomater 2019; 94:25-32. [PMID: 31226478 DOI: 10.1016/j.actbio.2019.06.020] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/03/2019] [Accepted: 06/13/2019] [Indexed: 01/09/2023]
Abstract
Following protein adsorption/activation which is the first step after the contact of material surfaces and whole blood (part 2), fibrinogen is converted to fibrin and platelets become activated and assembled in the form of a thrombus. This thrombus formation is the key feature that needs to be minimized in the creation of materials with low thrombogenicity. Further aspects of blood compatibility that are important on their own are complement and leukocyte activation which are also important drivers of thrombus formation. Hence this review summarizes the state of knowledge on all of these cascades and cells and their interactions. For each cascade or cell type, the chapter distinguishes statements which are in widespread agreement from statements where there is less of a consensus. STATEMENT OF SIGNIFICANCE: This paper is part 3 of a series of 4 reviews discussing the problem of biomaterial associated thrombogenicity. The objective was to highlight features of broad agreement and provide commentary on those aspects of the problem that were subject to dispute. We hope that future investigators will update these reviews as new scholarship resolves the uncertainties of today.
Collapse
Affiliation(s)
- Maud Gorbet
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Claudia Sperling
- Institute Biofunctional Polymer Materials, Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
| | - Manfred F Maitz
- Institute Biofunctional Polymer Materials, Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
| | - Christopher A Siedlecki
- Departments of Surgery and Bioengineering, The Pennsylvania State University, College of Medicine, Hershey, PA 17033, United States
| | - Carsten Werner
- Institute Biofunctional Polymer Materials, Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
| | - Michael V Sefton
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
41
|
Liu X, Gorzelanny C, Schneider SW. Platelets in Skin Autoimmune Diseases. Front Immunol 2019; 10:1453. [PMID: 31333641 PMCID: PMC6620619 DOI: 10.3389/fimmu.2019.01453] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE), systemic sclerosis (SSc), and small vessel vasculitis are three autoimmune diseases frequently manifested in the skin. They share common pathogenic features, including production of autoantibodies, loss of tolerance to self-antigens, tissue necrosis and fibrosis, vasculopathy and activation of the coagulation system. Platelets occupy a central part within the coagulation cascade and are well-recognized for their hemostatic role. However, recent cumulative evidence implicates their additional and multifaceted immunoregulatory functions. Platelets express immune receptors and they store growth factors, cytokines, and chemokines in their granules enabling a significant contribution to inflammation. A plethora of activating triggers such as damage associated molecular patterns (DAMPs) released from damaged endothelial cells, immune complexes, or complement effector molecules can mediate platelet activation. Activated platelets further foster an inflammatory environment and the crosstalk with the endothelium and leukocytes by the release of immunoactive molecules and microparticles. Further insight into the pathogenic implications of platelet activation will pave the way for new therapeutic strategies targeting autoimmune diseases. In this review, we discuss the inflammatory functions of platelets and their mechanistic contribution to the pathophysiology of SSc, ANCA associated small vessel vasculitis and other autoimmune diseases affecting the skin.
Collapse
Affiliation(s)
- Xiaobo Liu
- Department of Dermatology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Gorzelanny
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan W Schneider
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
42
|
Rijkers M, Schmidt D, Lu N, Kramer CSM, Heidt S, Mulder A, Porcelijn L, Claas FHJ, Leebeek FWG, Jansen AJG, Jongerius I, Zeerleder SS, Vidarsson G, Voorberg J, de Haas M. Anti-HLA antibodies with complementary and synergistic interaction geometries promote classical complement activation on platelets. Haematologica 2018; 104:403-416. [PMID: 30262558 PMCID: PMC6355480 DOI: 10.3324/haematol.2018.201665] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/19/2018] [Indexed: 12/21/2022] Open
Abstract
High titers of HLA antibodies are associated with platelet refractoriness, causing poor platelet increments after transfusions in a subset of patients with HLA antibodies. Currently, we do not know the biological mechanisms that explain the variability in clinical responses in HLA alloimmunized patients receiving platelet transfusions. Previously we showed that a subset of anti-HLA IgG-antibodies induces FcγRIIa-dependent platelet activation and enhanced phagocytosis. Here, we investigated whether anti-HLA IgG can induce complement activation on platelets. We found that a subset of anti-HLA IgG induced complement activation via the classical pathway, causing C4b and C3b deposition and formation of the membrane-attack complex. This resulted in permeabilization of platelet membranes and increased calcium influx. Complement activation also caused enhanced α-granule release, as measured by CD62P surface exposure. Blocking studies revealed that platelet activation was caused by FcγRIIa-dependent signaling as well as HLA antibody induced complement activation. Synergistic complement activation employing combinations of monoclonal IgGs suggested that assembly of oligomeric IgG complexes strongly promoted complement activation through binding of IgGs to different antigenic determinants on HLA. In agreement with this, we observed that preventing anti-HLA-IgG hexamer formation using an IgG-Fc:Fc blocking peptide, completely inhibited C3b and C4b deposition. Our results show that HLA antibodies can induce complement activation on platelets including membrane attack complex formation, pore formation and calcium influx. We propose that these events can contribute to fast platelet clearance in vivo in patients refractory to platelet transfusions with HLA alloantibodies, who may benefit from functional-platelet matching and treatment with complement inhibitors.
Collapse
Affiliation(s)
- Maaike Rijkers
- Department of Cellular and Molecular Hemostasis, Sanquin Research and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam
| | - David Schmidt
- Department of Experimental Immunohaematology, Sanquin Research and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam
| | - Nina Lu
- Department of Cellular and Molecular Hemostasis, Sanquin Research and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam
| | - Cynthia S M Kramer
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center
| | - Sebastiaan Heidt
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center
| | - Arend Mulder
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center
| | - Leendert Porcelijn
- Department of Immunohaematology Diagnostics, Sanquin Diagnostic Services, Amsterdam
| | - Frans H J Claas
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center
| | - Frank W G Leebeek
- Department of Hematology, Erasmus University Medical Center, Rotterdam
| | - A J Gerard Jansen
- Department of Cellular and Molecular Hemostasis, Sanquin Research and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam.,Department of Hematology, Erasmus University Medical Center, Rotterdam
| | - Ilse Jongerius
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam
| | - Sacha S Zeerleder
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam
| | - Gestur Vidarsson
- Department of Experimental Immunohaematology, Sanquin Research and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam
| | - Jan Voorberg
- Department of Cellular and Molecular Hemostasis, Sanquin Research and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam.,Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam
| | - Masja de Haas
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center .,Department of Immunohaematology Diagnostics, Sanquin Diagnostic Services, Amsterdam.,Center for Clinical Transfusion Research, Sanquin, Leiden, the Netherlands
| |
Collapse
|
43
|
Abbasian N, Herbert KE, Pawluczyk I, Burton JO, Bevington A. Vesicles bearing gifts: the functional importance of micro-RNA transfer in extracellular vesicles in chronic kidney disease. Am J Physiol Renal Physiol 2018; 315:F1430-F1443. [PMID: 30110570 DOI: 10.1152/ajprenal.00318.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), including microparticles (MPs) and exosomes (EXOs), are derived from a wide range of mammalian cells including blood platelets, endothelial cells, and kidney cells and can be detected in body fluids including blood and urine. While EVs are well established as diagnostic markers under pathophysiological and stress conditions, there is also mounting evidence of their functional significance as vehicles for communication between cells mediated by the presence of nucleic acids, especially microRNAs (miRs), encapsulated in the EVs. miRs regulate gene expression, are transported both in MPs and EXOs, and exert profound effects in the kidney. Here we review current understanding of the links between EVs and miRs, discuss the importance of miRs in kidney disease, and shed light on the role of EVs in transferring miRs through the circulation among the renal, vascular, and inflammatory cell populations that are functionally important in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Nima Abbasian
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| | - Karl E Herbert
- Department of Cardiovascular Sciences, University of Leicester, and Leicester National Institute of Health Research Cardiovascular Biomedical Research Unit , Leicester , United Kingdom
| | - Izabella Pawluczyk
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| | - James O Burton
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom.,John Walls Renal Unit, University Hospitals of Leicester , Leicester , United Kingdom
| | - Alan Bevington
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| |
Collapse
|
44
|
Complement links platelets to innate immunity. Semin Immunol 2018; 37:43-52. [DOI: 10.1016/j.smim.2018.01.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 12/11/2022]
|
45
|
Docheva N, Romero R, Chaemsaithong P, Tarca AL, Bhatti G, Pacora P, Panaitescu B, Chaiyasit N, Chaiworapongsa T, Maymon E, Hassan SS, Erez O. The profiles of soluble adhesion molecules in the "great obstetrical syndromes" . J Matern Fetal Neonatal Med 2018; 32:2113-2136. [PMID: 29320948 DOI: 10.1080/14767058.2018.1427058] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The objective of this study was to determine the profiles of maternal plasma soluble adhesion molecules in patients with preeclampsia, small-for-gestational-age (SGA) fetuses, acute pyelonephritis, preterm labor with intact membranes (PTL), preterm prelabor rupture of the membranes (preterm PROM), and fetal death. MATERIALS AND METHODS A cross-sectional study was conducted to determine maternal plasma concentrations of sE-selectin, sL-selectin, and sP-selectin as well as sICAM-1, sVCAM-1, and sPECAM-1 in patients with (1) an uncomplicated pregnancy (control, n = 100); (2) preeclampsia (n = 94); (3) SGA fetuses (in women without preeclampsia/hypertension, n = 45); (4) acute pyelonephritis (n = 25); (5) PTL (n = 53); (6) preterm PROM (n = 24); and (7) fetal death (n = 34). Concentrations of soluble adhesion molecules and inflammatory cytokines (tumor necrosis factor (TNF)-α and interleukin (IL)-8) were determined with sensitive and specific enzyme-linked immunoassays. RESULTS In comparison to women with a normal pregnancy, (1) women with preeclampsia had higher median concentrations of sE-selectin, sP-selectin, and sVCAM-1, and a lower concentration of sL-selectin (all p values < .001); (2) patients with SGA fetuses had higher median concentrations of sE-selectin, sP-selectin, and sVCAM-1 (all p values < .05); (3) patients with a fetal death had higher median concentrations of sE-selectin and sP-selectin (all p values < .05); (4) patients with acute pyelonephritis had higher median plasma concentrations of sE-selectin, sICAM-1, and sVCAM-1 (all p values < .001); (5) patients with preeclampsia and acute pyelonephritis, plasma concentrations of sVCAM-1, sE-selectin, and sP-selectin correlated with those of the proinflammatory cytokines TNF-α and interleukin (IL)-8 (all p values < .05); (6) patients with PTL had a higher median concentration of sP-selectin and a lower median concentration of VCAM-1 (all p values < .05); and (7) women with preterm PROM had lower median concentrations of sL-selectin and sVCAM-1 (all p values < .05). CONCLUSIONS The results of this study show that endothelial cell activation/dysfunction reflected by the plasma concentration of sE-selectin is not specific to preeclampsia but is present in pregnancies complicated by SGA fetuses, acute pyelonephritis, and fetal death. Collectively, we report that each obstetrical syndrome appears to have a stereotypical profile of soluble adhesion molecules in the peripheral circulation.
Collapse
Affiliation(s)
- Nikolina Docheva
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Roberto Romero
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,c Department of Obstetrics and Gynecology , University of Michigan , Ann Arbor , MI , USA.,d Department of Epidemiology and Biostatistics , Michigan State University , East Lansing , MI , USA.,e Center for Molecular Medicine and Genetics , Wayne State University , Detroit , MI , USA
| | - Piya Chaemsaithong
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Adi L Tarca
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Gaurav Bhatti
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Percy Pacora
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Bogdan Panaitescu
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Noppadol Chaiyasit
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Tinnakorn Chaiworapongsa
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Eli Maymon
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA.,f Department of Obstetrics and Gynecology , Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beersheba , Israel
| | - Sonia S Hassan
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA.,g Department of Physiology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Offer Erez
- a Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA.,f Department of Obstetrics and Gynecology , Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beersheba , Israel
| |
Collapse
|
46
|
|
47
|
Blatt AZ, Pathan S, Ferreira VP. Properdin: a tightly regulated critical inflammatory modulator. Immunol Rev 2017; 274:172-190. [PMID: 27782331 PMCID: PMC5096056 DOI: 10.1111/imr.12466] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The complement alternative pathway is a powerful arm of the innate immune system that enhances diverse inflammatory responses in the human host. Key to the effects of the alternative pathway is properdin, a serum glycoprotein that can both initiate and positively regulate alternative pathway activity. Properdin is produced by many different leukocyte subsets and circulates as cyclic oligomers of monomeric subunits. While the formation of non‐physiological aggregates in purified properdin preparations and the presence of potential properdin inhibitors in serum have complicated studies of its function, properdin has, regardless, emerged as a key player in various inflammatory disease models. Here, we review basic properdin biology, emphasizing the major hurdles that have complicated the interpretation of results from properdin‐centered studies. In addition, we elaborate on an emerging role for properdin in thromboinflammation and discuss the potential utility of properdin inhibitors as long‐term therapeutic options to treat diseases marked by increased formation of platelet/granulocyte aggregates. Finally, we describe the interplay between properdin and the alternative pathway negative regulator, Factor H, and how aiming to understand these interactions can provide scientists with the most effective ways to manipulate alternative pathway activation in complex systems.
Collapse
Affiliation(s)
- Adam Z Blatt
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sabina Pathan
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
| |
Collapse
|
48
|
Ekdahl KN, Teramura Y, Hamad OA, Asif S, Duehrkop C, Fromell K, Gustafson E, Hong J, Kozarcanin H, Magnusson PU, Huber-Lang M, Garred P, Nilsson B. Dangerous liaisons: complement, coagulation, and kallikrein/kinin cross-talk act as a linchpin in the events leading to thromboinflammation. Immunol Rev 2017; 274:245-269. [PMID: 27782319 DOI: 10.1111/imr.12471] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Innate immunity is fundamental to our defense against microorganisms. Physiologically, the intravascular innate immune system acts as a purging system that identifies and removes foreign substances leading to thromboinflammatory responses, tissue remodeling, and repair. It is also a key contributor to the adverse effects observed in many diseases and therapies involving biomaterials and therapeutic cells/organs. The intravascular innate immune system consists of the cascade systems of the blood (the complement, contact, coagulation, and fibrinolytic systems), the blood cells (polymorphonuclear cells, monocytes, platelets), and the endothelial cell lining of the vessels. Activation of the intravascular innate immune system in vivo leads to thromboinflammation that can be activated by several of the system's pathways and that initiates repair after tissue damage and leads to adverse reactions in several disorders and treatment modalities. In this review, we summarize the current knowledge in the field and discuss the obstacles that exist in order to study the cross-talk between the components of the intravascular innate immune system. These include the use of purified in vitro systems, animal models and various types of anticoagulants. In order to avoid some of these obstacles we have developed specialized human whole blood models that allow investigation of the cross-talk between the various cascade systems and the blood cells. We in particular stress that platelets are involved in these interactions and that the lectin pathway of the complement system is an emerging part of innate immunity that interacts with the contact/coagulation system. Understanding the resulting thromboinflammation will allow development of new therapeutic modalities.
Collapse
Affiliation(s)
- Kristina N Ekdahl
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden.,Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Yuji Teramura
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden.,Department of Bioengineering, The University of Tokyo, Tokyo, Japan
| | - Osama A Hamad
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Sana Asif
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Claudia Duehrkop
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Karin Fromell
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Elisabet Gustafson
- Department of Women's and Children's Health, Uppsala University Hospital, Uppsala, Sweden
| | - Jaan Hong
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Huda Kozarcanin
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Markus Huber-Lang
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University of Ulm, Ulm, Germany
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Faculty of Health and Medical Sciences, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
49
|
Distinct contributions of complement factors to platelet activation and fibrin formation in venous thrombus development. Blood 2017; 129:2291-2302. [PMID: 28223279 DOI: 10.1182/blood-2016-11-749879] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 02/07/2017] [Indexed: 12/30/2022] Open
Abstract
Expanding evidence indicates multiple interactions between the hemostatic system and innate immunity, and the coagulation and complement cascades. Here we show in a tissue factor (TF)-dependent model of flow restriction-induced venous thrombosis that complement factors make distinct contributions to platelet activation and fibrin deposition. Complement factor 3 (C3) deficiency causes prolonged bleeding, reduced thrombus incidence, thrombus size, fibrin and platelet deposition in the ligated inferior vena cava, and diminished platelet activation in vitro. Initial fibrin deposition at the vessel wall over 6 hours in this model was dependent on protein disulfide isomerase (PDI) and TF expression by myeloid cells, but did not require neutrophil extracellular trap formation involving peptidyl arginine deiminase 4. In contrast to C3-/- mice, C5-deficient mice had no apparent defect in platelet activation in vitro, and vessel wall platelet deposition and initial hemostasis in vivo. However, fibrin formation, the exposure of negatively charged phosphatidylserine (PS) on adherent leukocytes, and clot burden after 48 hours were significantly reduced in C5-/- mice compared with wild-type controls. These results delineate that C3 plays specific roles in platelet activation independent of formation of the terminal complement complex and provide in vivo evidence for contributions of complement-dependent membrane perturbations to prothrombotic TF activation on myeloid cells.
Collapse
|
50
|
Ferluga J, Kouser L, Murugaiah V, Sim RB, Kishore U. Potential influences of complement factor H in autoimmune inflammatory and thrombotic disorders. Mol Immunol 2017; 84:84-106. [PMID: 28216098 DOI: 10.1016/j.molimm.2017.01.015] [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] [Received: 08/26/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 01/01/2023]
Abstract
Complement system homeostasis is important for host self-protection and anti-microbial immune surveillance, and recent research indicates roles in tissue development and remodelling. Complement also appears to have several points of interaction with the blood coagulation system. Deficiency and altered function due to gene mutations and polymorphisms in complement effectors and regulators, including Factor H, have been associated with familial and sporadic autoimmune inflammatory - thrombotic disorders, in which autoantibodies play a part. These include systemic lupus erythematosus, rheumatoid arthritis, atypical haemolytic uremic syndrome, anti-phospholipid syndrome and age-related macular degeneration. Such diseases are generally complex - multigenic and heterogeneous in their symptoms and predisposition/susceptibility. They usually need to be triggered by vascular trauma, drugs or infection and non-complement genetic factors also play a part. Underlying events seem to include decline in peripheral regulatory T cells, dendritic cell, and B cell tolerance, associated with alterations in lymphoid organ microenvironment. Factor H is an abundant protein, synthesised in many cell types, and its reported binding to many different ligands, even if not of high affinity, may influence a large number of molecular interactions, together with the accepted role of Factor H within the complement system. Factor H is involved in mesenchymal stem cell mediated tolerance and also contributes to self-tolerance by augmenting iC3b production and opsonisation of apoptotic cells for their silent dendritic cell engulfment via complement receptor CR3, which mediates anti-inflammatory-tolerogenic effects in the apoptotic cell context. There may be co-operation with other phagocytic receptors, such as complement C1q receptors, and the Tim glycoprotein family, which specifically bind phosphatidylserine expressed on the apoptotic cell surface. Factor H is able to discriminate between self and nonself surfaces for self-protection and anti-microbe defence. Factor H, particularly as an abundant platelet protein, may also modulate blood coagulation, having an anti-thrombotic role. Here, we review a number of interaction pathways in coagulation and in immunity, together with associated diseases, and indicate where Factor H may be expected to exert an influence, based on reports of the diversity of ligands for Factor H.
Collapse
Affiliation(s)
- Janez Ferluga
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, United Kingdom
| | - Lubna Kouser
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, United Kingdom
| | - Valarmathy Murugaiah
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, United Kingdom
| | - Robert B Sim
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, United Kingdom.
| |
Collapse
|