1
|
Jiang K, Jia Y, Chen L, Huang F, Chen M. Association of Interleukin-17 Inhibitors With Hypertension in Patients With Autoimmune Diseases: A Systematic Review and Meta-analysis on Randomized Controlled Trials. J Cardiovasc Pharmacol 2024; 83:557-564. [PMID: 38417026 DOI: 10.1097/fjc.0000000000001547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/21/2024] [Indexed: 03/01/2024]
Abstract
Abstract:
The influence of interleukin (IL)-17 inhibition on blood pressure in patients with autoimmune diseases remains inconclusive. Our objective is to examine the risk of hypertension in patients with autoimmune diseases undergoing IL-17 inhibition therapies through meta-analysis of randomized, placebo-controlled trials. We obtained integrated data from PubMed, Embase, and ClinicalTrials.gov. Incident hypertension rates were calculated, and hazard ratios with 95% confidence intervals were analyzed, along with
statistics to assess heterogeneity. Sequential analysis ensured conclusion reliability. In 30 randomized controlled trials involving 9909 patients with diverse autoimmune diseases treated with anti-IL-17 agents, our meta-analysis revealed a significant increase in hypertension risk (risk ratio 1.69, 95% confidence interval 1.24–2.31, P = 0.001), robustly supported by trial sequential analysis. Among the 4 agents (secukinumab, ixekizumab, bimekizumab, and brodalumab), only secukinumab exhibited a notable association with hypertension. Patients with various primary autoimmune diseases, particularly those with psoriatic arthritis, had a higher likelihood of developing hypertension; in rheumatic arthritis patient cohorts, anti-IL-17 agents did not elevate hypertension risk. Prolonged treatment duration correlated with an increased hypertension risk. Stratifying by sex, studies with a female predominance demonstrated a higher risk ratio for hypertension compared with male-predominant studies. This highlights that anti-IL-17 treatment escalates hypertension risk, emphasizing the need for extra caution when managing patients with autoimmune diseases (Registered by PROSPERO, CRD42016053112).
Collapse
Affiliation(s)
- Kexin Jiang
- Department of Cardiology
- Laboratory of Heart Valve Disease; and
| | - Yuheng Jia
- Department of Cardiology
- Laboratory of Heart Valve Disease; and
| | - Li Chen
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fangyang Huang
- Department of Cardiology
- Laboratory of Heart Valve Disease; and
| | - Mao Chen
- Department of Cardiology
- Laboratory of Heart Valve Disease; and
| |
Collapse
|
2
|
Huang M, Wang L, Zhang Q, Zhou L, Liao R, Wu A, Wang X, Luo J, Huang F, Zou W, Wu J. Interleukins in Platelet Biology: Unraveling the Complex Regulatory Network. Pharmaceuticals (Basel) 2024; 17:109. [PMID: 38256942 PMCID: PMC10820339 DOI: 10.3390/ph17010109] [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: 12/20/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Interleukins, a diverse family of cytokines produced by various cells, play crucial roles in immune responses, immunoregulation, and a wide range of physiological and pathological processes. In the context of megakaryopoiesis, thrombopoiesis, and platelet function, interleukins have emerged as key regulators, exerting significant influence on the development, maturation, and activity of megakaryocytes (MKs) and platelets. While the therapeutic potential of interleukins in platelet-related diseases has been recognized for decades, their clinical application has been hindered by limitations in basic research and challenges in drug development. Recent advancements in understanding the molecular mechanisms of interleukins and their interactions with MKs and platelets, coupled with breakthroughs in cytokine engineering, have revitalized the field of interleukin-based therapeutics. These breakthroughs have paved the way for the development of more effective and specific interleukin-based therapies for the treatment of platelet disorders. This review provides a comprehensive overview of the effects of interleukins on megakaryopoiesis, thrombopoiesis, and platelet function. It highlights the potential clinical applications of interleukins in regulating megakaryopoiesis and platelet function and discusses the latest bioengineering technologies that could improve the pharmacokinetic properties of interleukins. By synthesizing the current knowledge in this field, this review aims to provide valuable insights for future research into the clinical application of interleukins in platelet-related diseases.
Collapse
Affiliation(s)
- Miao Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (M.H.); (Q.Z.)
| | - Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.W.); (L.Z.); (R.L.); (A.W.); (F.H.)
| | - Qianhui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (M.H.); (Q.Z.)
| | - Ling Zhou
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.W.); (L.Z.); (R.L.); (A.W.); (F.H.)
| | - Rui Liao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.W.); (L.Z.); (R.L.); (A.W.); (F.H.)
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.W.); (L.Z.); (R.L.); (A.W.); (F.H.)
| | - Xinle Wang
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (X.W.); (J.L.)
| | - Jiesi Luo
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (X.W.); (J.L.)
| | - Feihong Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.W.); (L.Z.); (R.L.); (A.W.); (F.H.)
| | - Wenjun Zou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (M.H.); (Q.Z.)
| | - Jianming Wu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (X.W.); (J.L.)
- The Key Laboratory of Medical Electrophysiology, Institute of Cardiovascular Research, Ministry of Education of China, Luzhou 646000, China
| |
Collapse
|
3
|
Jiang Z, Jiang X, Chen A, He W. Platelet activation: a promoter for psoriasis and its comorbidity, cardiovascular disease. Front Immunol 2023; 14:1238647. [PMID: 37654493 PMCID: PMC10465348 DOI: 10.3389/fimmu.2023.1238647] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease with a prevalence of 0.14% to 1.99%. The underlying pathology is mainly driven by the abnormal immune responses including activation of Th1, Th17, Th22 cells and secretion of cytokines. Patients with psoriasis are more likely to develop cardiovascular disease (CVD) which has been well recognized as a comorbidity of psoriasis. As mediators of hemostasis and thromboinflammation, platelets play an important part in CVD. However, less is known about their pathophysiological contribution to psoriasis and psoriasis-associated CVD. A comprehensive understanding of the role of platelet activation in psoriasis might pave the path for more accurate prediction of cardiovascular (CV) risk and provide new strategies for psoriasis management, which alleviates the increased CV burden associated with psoriasis. Here we review the available evidence about the biomarkers and mechanisms of platelet activation in psoriasis and the role of platelet activation in intriguing the common comorbidity, CVD. We further discussed the implications and efficacy of antiplatelet therapies in the treatment of psoriasis and prevention of psoriasis-associated CVD.
Collapse
Affiliation(s)
- Ziqi Jiang
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoran Jiang
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Aijun Chen
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenyan He
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Vellecco V, Saviano A, Raucci F, Casillo GM, Mansour AA, Panza E, Mitidieri E, Femminella GD, Ferrara N, Cirino G, Sorrentino R, Iqbal AJ, d'Emmanuele di Villa Bianca R, Bucci M, Maione F. Interleukin-17 (IL-17) triggers systemic inflammation, peripheral vascular dysfunction, and related prothrombotic state in a mouse model of Alzheimer's disease. Pharmacol Res 2023; 187:106595. [PMID: 36470548 DOI: 10.1016/j.phrs.2022.106595] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/08/2022]
Abstract
Alzheimer's disease (AD) is one of the most prevalent forms of neurodegenerative disorders. Previously, we have shown that in vivo administration of an IL-17 neutralizing antibody (IL-17Ab) rescues amyloid-β-induced neuro-inflammation and memory impairment, demonstrating the pivotal role of IL-17 in AD-derived cognitive deficit. Recently, AD has been recognized as a more intriguing pathology affecting vascular networks and platelet function. However, not much is known about peripheral vascular inflammation and how pro-inflammatory circulating cells/mediators could affect peripheral vessels' function. This study aimed to evaluate whether IL-17Ab treatment could also impact peripheral AD features, such as systemic inflammation, peripheral vascular dysfunction, and related pro-thrombotic state in a non-genetic mouse model of AD. Mice were injected intracerebroventricularly with Aβ1-42 peptide (3 μg/3 μl). To evaluate the systemic/peripheral protective profile of IL-17Ab, we used an intranasal administration of IL-17Ab (1 μg/10 μl) at 5, 12, and 19 days after Aβ1-42 injection. Circulating Th17/Treg cells and related cyto-chemokines, haematological parameters, vascular/endothelial reactivity, platelets and coagulation function in mice were evaluated. IL-17Ab treatment ameliorates the systemic/peripheral inflammation, immunological perturbance, vascular/endothelial impairment and pro-thrombotic state, suggesting a key role for this cytokine in fostering inflammatory processes that characterize the multifaced aspects of AD.
Collapse
Affiliation(s)
- Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Elisabetta Panza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Grazia Daniela Femminella
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy.
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; Istituti Clinici Scientifici ICS-Maugeri, Telese Terme, BN, Italy.
| | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnologies, School of Medicine, University of Naples, Federico II, Via Pansini, 5, 80131 Naples, Italy.
| | - Asif Jilani Iqbal
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | | | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| |
Collapse
|
5
|
The Association of Patent Ductus Arteriosus with Inflammation: A Narrative Review of the Role of Inflammatory Biomarkers and Treatment Strategy in Premature Infants. Int J Mol Sci 2022; 23:ijms232213877. [PMID: 36430355 PMCID: PMC9699120 DOI: 10.3390/ijms232213877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/27/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Patent ductus arteriosus (PDA) is a common cardiovascular complication that complicates clinical care in the intensive care of premature infants. Prenatal and postnatal infections and the inflammation process can contribute to PDA, and intrauterine inflammation is a known risk factor of PDA. A variety of inflammatory biomarkers have been reported to be associated with PDA. Chorioamnionitis induces the fetal inflammatory process via several cytokines that have been reported to be associated with the presence of PDA and may have a role in the vascular remodeling process or vessel dilation of the ductus. On the other hand, anti-inflammatory agents, such as antenatal steroids, decrease PDA incidence and severity in patients born to those with chorioamnionitis. Proinflammatory cytokines, which are expressed more significantly in preterm neonates and chorioamnionitis, are associated with the presence of PDA. In this review, we focus on the pathogenesis of PDA in preterm infants and the role of biomarkers associated with the perinatal inflammatory process.
Collapse
|
6
|
Resende GG, da Cruz Lage R, Lobê SQ, Medeiros AF, Costa E Silva AD, Nogueira Sá AT, Oliveira AJDA, Sousa D, Guimarães HC, Gomes IC, Souza RP, Aguiar RS, Tunala R, Forestiero F, Bueno Filho JSS, Teixeira MM. Blockade of interleukin seventeen (IL-17A) with secukinumab in hospitalized COVID-19 patients - the BISHOP study. Infect Dis (Lond) 2022; 54:591-599. [PMID: 35485381 DOI: 10.1080/23744235.2022.2066171] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Patients with severe COVID-19 seem to evolve with a compromised antiviral response and hyperinflammation. Neutrophils are critical players in COVID-19. IL-17A plays a major role in protection against extracellular pathogens and neutrophil attraction/activation. We hypothesized that secukinumab, an anti-IL17A monoclonal antibody, could prevent the deleterious hyperinflammation in COVID-19. METHODS BISHOP was a randomized, open-label, single-centre, phase-II controlled trial. Fifty adult patients hospitalized with PCR-positive Covid-19, were randomized 1:1 to receive 300 mg of secukinumab subcutaneously at day-0 plus standard of care (group A) or standard of care alone (group B). A second dose of 300 mg of secukinumab could be administered on day-7, according to staff judgement. The primary endpoint was ventilator-free days at day-28 (VFD-28). Secondary efficacy and safety outcomes were also explored. RESULTS An intention-to-treat analysis showed no difference in VFD-28: 23.7 (95%CI 19.6-27.8) in group A vs. 23.8 (19.9-27.6) in group B, p = .62; There was also no difference in hospitalization time, intensive care unit demand and the incidence of circulatory shock, acute kidney injury, fungal or bacterial co-infections. There was no difference in the incidence of severe adverse events. Pulmonary thromboembolism occurred only in males and was less frequent in secukinumab-treated patients (4.2% vs. 26.2% p = .04). There was one death in each group. Upper airway viral clearance was also similar in both groups. CONCLUSION The efficacy of secukinumab in the treatment of Covid19 was not demonstrated. Secukinumab decreased pulmonary embolism in male patients. There was no difference between groups in adverse events and no unexpected events were observed.
Collapse
Affiliation(s)
- Gustavo Gomes Resende
- Rheumatology Unit, Hospital das Clínicas - Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ricardo da Cruz Lage
- Rheumatology Unit, Hospital das Clínicas - Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | | | | | | | | | - Denise Sousa
- Hospital Risoleta Tolentino Neves, Belo Horizonte, Brazil
| | | | | | - Renan Pedra Souza
- Dept. of Genetics, Ecology and Evolution - UFMG, Belo Horizonte, Brazil
| | | | | | | | | | | |
Collapse
|
7
|
Dalbeni A, Castelli M, Zoncapè M, Minuz P, Sacerdoti D. Platelets in Non-alcoholic Fatty Liver Disease. Front Pharmacol 2022; 13:842636. [PMID: 35250588 PMCID: PMC8895200 DOI: 10.3389/fphar.2022.842636] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/03/2022] [Indexed: 12/17/2022] Open
Abstract
Non alcoholic steatohepatitis (NASH) is the inflammatory reaction of the liver to excessive accumulation of lipids in the hepatocytes. NASH can progress to cirrhosis and hepatocellular carcinoma (HCC). Fatty liver is the hepatic manifestation of metabolic syndrome. A subclinical inflammatory state is present in patients with metabolic alterations like insulin resistance, type-2 diabetes, obesity, hyperlipidemia, and hypertension. Platelets participate in immune cells recruitment and cytokines-induced liver damage. It is hypothesized that lipid toxicity cause accumulation of platelets in the liver, platelet adhesion and activation, which primes the immunoinflammatory reaction and activation of stellate cells. Recent data suggest that antiplatelet drugs may interrupt this cascade and prevent/improve NASH. They may also improve some metabolic alterations. The pathophysiology of inflammatory liver disease and the implication of platelets are discussed in details.
Collapse
Affiliation(s)
- Andrea Dalbeni
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Marco Castelli
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Mirko Zoncapè
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Pietro Minuz
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- *Correspondence: Pietro Minuz,
| | - David Sacerdoti
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| |
Collapse
|
8
|
Shavit-Stein E, Berkowitz S, Gofrit SG, Altman K, Weinberg N, Maggio N. Neurocoagulation from a Mechanistic Point of View in the Central Nervous System. Semin Thromb Hemost 2022; 48:277-287. [PMID: 35052009 DOI: 10.1055/s-0041-1741569] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coagulation mechanisms are critical for maintaining homeostasis in the central nervous system (CNS). Thrombin, an important player of the coagulation cascade, activates protease activator receptors (PARs), members of the G-protein coupled receptor family. PAR1 is located on neurons and glia. Following thrombin activation, PAR1 signals through the extracellular signal-regulated kinase pathway, causing alterations in neuronal glutamate release and astrocytic morphological changes. Similarly, the anticoagulation factor activated protein C (aPC) can cleave PAR1, following interaction with the endothelial protein C receptor. Both thrombin and aPC are expressed on endothelial cells and pericytes in the blood-brain barrier (BBB). Thrombin-induced PAR1 activation increases cytosolic Ca2+ concentration in brain vessels, resulting in nitric oxide release and increasing F-actin stress fibers, damaging BBB integrity. aPC also induces PAR1 activation and preserves BBB vascular integrity via coupling to sphingosine 1 phosphate receptors. Thrombin-induced PAR1 overactivation and BBB disruption are evident in CNS pathologies. During epileptic seizures, BBB disruption promotes thrombin penetration. Thrombin induces PAR1 activation and potentiates N-methyl-D-aspartate receptors, inducing glutamate-mediated hyperexcitability. Specific PAR1 inhibition decreases status epilepticus severity in vivo. In stroke, the elevation of brain thrombin levels further compromises BBB integrity, with direct parenchymal damage, while systemic factor Xa inhibition improves neurological outcomes. In multiple sclerosis (MS), brain thrombin inhibitory capacity correlates with clinical presentation. Both thrombin inhibition by hirudin and the use of recombinant aPC improve disease severity in an MS animal model. This review presents the mechanisms underlying the effects of coagulation on the physiology and pathophysiology of the CNS.
Collapse
Affiliation(s)
- Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shani Berkowitz
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shany Guly Gofrit
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Keren Altman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Nitai Weinberg
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan, Israel
| |
Collapse
|
9
|
Johnson BZ, Stevenson AW, Barrett LW, Fear MW, Wood FM, Linden MD. Platelets after burn injury - hemostasis and beyond. Platelets 2022; 33:655-665. [PMID: 34986759 DOI: 10.1080/09537104.2021.1981849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Burn injuries are common and often life-threatening trauma. With this trauma comes an interruption of normal hemostasis, with distinct impacts on platelets. Our interest in the relationships between burn injury and platelet function stems from two key perspectives: platelet function is a vital component of acute responses to injury, and furthermore the incidence of cardiovascular disease (CVD) is higher in burn survivors compared to the general population. This review explores the impact of burn injury on coagulation, platelet function, and the participation of platelets in immunopathology. Potential avenues of further research are explored, and consideration is given to what therapies may be appropriate for mediating post-burn thrombopathology.
Collapse
Affiliation(s)
- B Z Johnson
- Burn Injury Research Unit, University of Western Australia, Perth, Australia.,School of Biomedical Science, University of Western Australia, Perth, Australia
| | - A W Stevenson
- Burn Injury Research Unit, University of Western Australia, Perth, Australia.,School of Biomedical Science, University of Western Australia, Perth, Australia
| | - L W Barrett
- Burn Injury Research Unit, University of Western Australia, Perth, Australia.,Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - M W Fear
- Burn Injury Research Unit, University of Western Australia, Perth, Australia.,School of Biomedical Science, University of Western Australia, Perth, Australia
| | - F M Wood
- Burn Injury Research Unit, University of Western Australia, Perth, Australia.,Burns Service of Western Australia, Wa Department of Health, Nedlands, Australia
| | - M D Linden
- School of Biomedical Science, University of Western Australia, Perth, Australia
| |
Collapse
|
10
|
Page MJ, Pretorius E. Platelet Behavior Contributes to Neuropathologies: A Focus on Alzheimer's and Parkinson's Disease. Semin Thromb Hemost 2021; 48:382-404. [PMID: 34624913 DOI: 10.1055/s-0041-1733960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The functions of platelets are broad. Platelets function in hemostasis and thrombosis, inflammation and immune responses, vascular regulation, and host defense against invading pathogens, among others. These actions are achieved through the release of a wide set of coagulative, vascular, inflammatory, and other factors as well as diverse cell surface receptors involved in the same activities. As active participants in these physiological processes, platelets become involved in signaling pathways and pathological reactions that contribute to diseases that are defined by inflammation (including by pathogen-derived stimuli), vascular dysfunction, and coagulation. These diseases include Alzheimer's and Parkinson's disease, the two most common neurodegenerative diseases. Despite their unique pathological and clinical features, significant shared pathological processes exist between these two conditions, particularly relating to a central inflammatory mechanism involving both neuroinflammation and inflammation in the systemic environment, but also neurovascular dysfunction and coagulopathy, processes which also share initiation factors and receptors. This triad of dysfunction-(neuro)inflammation, neurovascular dysfunction, and hypercoagulation-illustrates the important roles platelets play in neuropathology. Although some mechanisms are understudied in Alzheimer's and Parkinson's disease, a strong case can be made for the relevance of platelets in neurodegeneration-related processes.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| |
Collapse
|
11
|
Maione F, Casillo GM, Raucci F, Salvatore C, Ambrosini G, Costa L, Scarpa R, Caso F, Bucci M. Interleukin-17A (IL-17A): A silent amplifier of COVID-19. Biomed Pharmacother 2021; 142:111980. [PMID: 34364043 PMCID: PMC8318692 DOI: 10.1016/j.biopha.2021.111980] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 12/27/2022] Open
Abstract
One of the hallmarks of COVID-19 is the cytokine storm that provokes primarily pneumonia followed by systemic inflammation. Emerging evidence has identified a potential link between elevated interleukin-17A (IL-17A) levels and disease severity and progression. Considering that per se, IL-17A can activate several inflammatory pathways, it is plausible to hypothesize an involvement of this cytokine in COVID-19 clinical outcomes. Thus, IL-17A could represent a marker of disease progression and/or a target to develop therapeutic strategies. This hypothesis paper aims to propose this "unique" cytokine as a silent amplifier of the COVID-19 immune response and (potentially) related therapy.
Collapse
Affiliation(s)
- Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Federica Raucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Cristian Salvatore
- New.Fa.DEm SRL, Viale Ferrovie Dello Stato, 1, 80014 Giugliano in Campania, Naples, Italy
| | - Giovanna Ambrosini
- New.Fa.DEm SRL, Viale Ferrovie Dello Stato, 1, 80014 Giugliano in Campania, Naples, Italy
| | - Luisa Costa
- Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Raffaele Scarpa
- Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Francesco Caso
- Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini, 5, 80131 Naples, Italy.
| | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| |
Collapse
|
12
|
Gatsiou A, Sopova K, Tselepis A, Stellos K. Interleukin-17A Triggers the Release of Platelet-Derived Factors Driving Vascular Endothelial Cells toward a Pro-Angiogenic State. Cells 2021; 10:1855. [PMID: 34440624 PMCID: PMC8392697 DOI: 10.3390/cells10081855] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 01/26/2023] Open
Abstract
Platelets comprise a highly interactive immune cell subset of the circulatory system traditionally known for their unique haemostatic properties. Although platelets are considered as a vault of growth factors, cytokines and chemokines with pivotal role in vascular regeneration and angiogenesis, the exact mechanisms by which they influence vascular endothelial cells (ECs) function remain underappreciated. In the present study, we examined the role of human IL-17A/IL-17RA axis in platelet-mediated pro-angiogenic responses. We reveal that IL-17A receptor (IL-17RA) mRNA is present in platelets transcriptome and a profound increase is documented on the surface of activated platelets. By quantifying the protein levels of several factors, involved in angiogenesis, we identified that IL-17A/IL17RA axis selectively induces the release of vascular endothelial growth factor, interleukin -2 and -4, as well as monocyte chemoattractant protein -1 from treated platelets. However, IL-17A exerted no effect on the release of IL-10, an anti-inflammatory factor with potentially anti-angiogenic properties, from platelets. Treatment of human endothelial cell two-dimensional tubule networks or three-dimensional spheroid and mouse aortic ring structures with IL-17A-induced platelet releasate evoked pro-angiogenic responses of ECs. Our findings suggest that IL-17A may critically affect platelet release of pro-angiogenic factors driving ECs towards a pro-angiogenic state.
Collapse
Affiliation(s)
- Aikaterini Gatsiou
- RNA Metabolism and Vascular Inflammation Group, Center of Molecular Medicine, Institute of Cardiovascular Regeneration, Johann Wolfgang Goethe University, 60596 Frankfurt am Main, Germany; (A.G.); (K.S.)
- Laboratory of Clinical Biochemistry, Atherothrombosis Research Center, University of Ioannina, 45110 Ioannina, Greece;
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK
| | - Kateryna Sopova
- RNA Metabolism and Vascular Inflammation Group, Center of Molecular Medicine, Institute of Cardiovascular Regeneration, Johann Wolfgang Goethe University, 60596 Frankfurt am Main, Germany; (A.G.); (K.S.)
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK
- Department of Cardiology, Freeman Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Alexandros Tselepis
- Laboratory of Clinical Biochemistry, Atherothrombosis Research Center, University of Ioannina, 45110 Ioannina, Greece;
| | - Konstantinos Stellos
- RNA Metabolism and Vascular Inflammation Group, Center of Molecular Medicine, Institute of Cardiovascular Regeneration, Johann Wolfgang Goethe University, 60596 Frankfurt am Main, Germany; (A.G.); (K.S.)
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK
- Department of Cardiology, Freeman Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| |
Collapse
|
13
|
Visser MJE, Tarr G, Pretorius E. Thrombosis in Psoriasis: Cutaneous Cytokine Production as a Potential Driving Force of Haemostatic Dysregulation and Subsequent Cardiovascular Risk. Front Immunol 2021; 12:688861. [PMID: 34335591 PMCID: PMC8324086 DOI: 10.3389/fimmu.2021.688861] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/06/2021] [Indexed: 12/18/2022] Open
Abstract
Psoriasis (PsO) is a common T cell-mediated inflammatory disorder of the skin with an estimated prevalence of 2%. The condition manifests most commonly as erythematous plaques covered with scales. The aetiology of PsO is multifactorial and disease initiation involves interactions between environmental factors, susceptibility genes, and innate and adaptive immune responses. The underlying pathology is mainly driven by interleukin-17. In addition, various inflammatory mediators from specific T helper (TH) cell subsets, namely TH1, TH17, and TH22, are overexpressed in cutaneous lesions and may also be detected in the peripheral blood of psoriatic patients. Moreover, these individuals are also at greater risk, compared to the general population, of developing multiple comorbid conditions. Cardiovascular disease (CVD) has been recognised as a prominent comorbidity of PsO. A potential mechanism contributing to this association may be the presence of a hypercoagulable state in these individuals. Inflammation and coagulation are closely related. The presence of chronic, low-grade systemic inflammation may promote thrombosis – one of the major determinants of CVD. A pro-inflammatory milieu may induce the expression of tissue factor, augment platelet activity, and perturb the vascular endothelium. Altogether, these changes will result in a prothrombotic state. In this review, we describe the aetiology of PsO, as well as the pathophysiology of the condition. We also consider its relationship to CVD. Given the systemic inflammatory nature of PsO, we evaluate the potential contribution of prominent inflammatory mediators (implicated in PsO pathogenesis) to establishing a prothrombotic state in psoriatic patients.
Collapse
Affiliation(s)
- Maria J E Visser
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Gareth Tarr
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,Division of Rheumatology, Institute of Orthopaedics and Rheumatology, Winelands Mediclinic Orthopaedic Hospital, Stellenbosch, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
14
|
Gromadziński L, Paukszto Ł, Skowrońska A, Holak P, Smoliński M, Łopieńska-Biernat E, Lepiarczyk E, Lipka A, Jastrzębski JP, Majewska M. Transcriptomic Profiling of Femoral Veins in Deep Vein Thrombosis in a Porcine Model. Cells 2021; 10:1576. [PMID: 34206566 PMCID: PMC8304794 DOI: 10.3390/cells10071576] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/13/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022] Open
Abstract
Deep vein thrombosis (DVT) is a severe disease affecting the human venous system, accompanied by high morbidity and mortality rates caused by early and late complications. The study aimed at analyzing the changes in the transcriptome of the femoral vein caused by DVT in the porcine model based on the formation of the thrombus in vivo. The study was performed on 11 castrated male pigs: A thrombus was formed in each left femoral vein in six animals; the remaining five served as a control group. Total RNA was isolated from the left femoral veins of the experimental and control animals. High-throughput RNA sequencing was used to analyze the global changes in the transcriptome of veins with induced DVT. Applied multistep bioinformatics revealed 1474 differentially expressed genes (DEGs): 1019 upregulated and 455 downregulated. Functional Gene Ontology annotated 1220 of DEGs into 225 biological processes, 30 molecular functions and 40 cellular components categories. KEGG analysis disclosed TNF, NF-κB and apoptosis pathways' overexpression in DVT samples. A thorough analysis of the detected DEGs indicated that a dysregulated inflammatory response and disturbed balance between clotting and anti-clotting factors play a crucial role in the process of DVT.
Collapse
Affiliation(s)
- Leszek Gromadziński
- Department of Cardiology and Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Łukasz Paukszto
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland; (Ł.P.); (J.P.J.)
| | - Agnieszka Skowrońska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland; (A.S.); (E.L.)
| | - Piotr Holak
- Department of Surgery and Radiology with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Michał Smoliński
- Clinic of Cardiology and Internal Diseases, University Clinical Hospital in Olsztyn, 10-082 Olsztyn, Poland;
| | - Elżbieta Łopieńska-Biernat
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Ewa Lepiarczyk
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland; (A.S.); (E.L.)
| | - Aleksandra Lipka
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland;
| | - Jan Paweł Jastrzębski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland; (Ł.P.); (J.P.J.)
| | - Marta Majewska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland; (A.S.); (E.L.)
| |
Collapse
|
15
|
Herster F, Karbach S, Chatterjee M, Weber ANR. Platelets: Underestimated Regulators of Autoinflammation in Psoriasis. J Invest Dermatol 2021; 141:1395-1403. [PMID: 33810836 DOI: 10.1016/j.jid.2020.12.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 01/01/2023]
Abstract
Platelets have long been known as mediators of hemostasis and, more recently, as mediators of thromboinflammation, although their physiopathological role has mostly been investigated in the context of disease of internal organs, such as liver and kidney, or systemic disorders. Of late, exciting recent data suggest that platelets may also play a role in inflammation at distal sites such as the skin: recent studies show that platelets, by engaging polymorphonuclear neutrophils (PMNs), contribute to local inflammation in the frequent skin disorder, psoriasis. In an experimental model, systemic depletion of platelets drastically attenuated skin inflammation by preventing PMN infiltration of the skin. A broader role of platelets in different types of skin inflammation is therefore likely, and in this paper, we specifically review recent advances in psoriasis. Special emphasis is given to the crosstalk with systemic platelet effects, which may be of interest in psoriasis-related cardiovascular comorbidities. Furthermore, we discuss the potential for platelet-centered interventions in the therapy for psoriasis.
Collapse
Affiliation(s)
- Franziska Herster
- Department of Immunology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany; Department of Molecular Oncology, Robert Bosch Centrum für Tumorerkrankungen (RBCT), Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Susanne Karbach
- Center for Cardiology - Cardiology I, University Medical Center Mainz and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Madhumita Chatterjee
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- Department of Immunology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
16
|
Sabioni L, De Lorenzo A, Lamas C, Muccillo F, Castro-Faria-Neto HC, Estato V, Tibirica E. Systemic microvascular endothelial dysfunction and disease severity in COVID-19 patients: Evaluation by laser Doppler perfusion monitoring and cytokine/chemokine analysis. Microvasc Res 2021; 134:104119. [PMID: 33278457 PMCID: PMC7710468 DOI: 10.1016/j.mvr.2020.104119] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Microvascular dysfunction, serum cytokines and chemokines may play important roles in pathophysiology of coronavirus disease 2019 (COVID-19), especially in severe cases. METHODS Patients with COVID-19 underwent non-invasive evaluation of systemic endothelium-dependent microvascular reactivity - using laser Doppler perfusion monitoring in the skin of the forearm - coupled to local thermal hyperemia. Maximal microvascular vasodilatation (44 °C thermal plateau phase) was used as endpoint. A multiplex biometric immunoassay was used to assess a panel of 48 serum cytokines and chemokines. Severe COVID-19 (S-COVID) was defined according to WHO criteria, while all other cases of COVID-19 were considered mild to moderate (M-COVID). A group of healthy individuals who tested negative for SARS-CoV-2 served as a control group and was also evaluated with LDPM. RESULTS Thirty-two patients with COVID-19 (25% S-COVID) and 14 controls were included. Basal microvascular flow was similar between M-COVID and controls (P = 0.69) but was higher in S-COVID than in controls (P = 0.005) and M-COVID patients (P = 0.01). The peak microvascular vasodilator response was markedly decreased in both patient groups (M-COVID, P = 0.001; S-COVID, P < 0.0001) compared to the healthy group. The percent increases in microvascular flow were markedly reduced in both patient groups (M-COVID, P < 0.0001; S-COVID, P < 0.0001) compared to controls. Patients with S-COVID had markedly higher concentrations of dissimilar proinflammatory cytokines and chemokines, compared to patients with M-COVID. CONCLUSIONS In patients with COVID-19, especially with S-COVID, endothelium-dependent microvascular vasodilator responses are reduced, while serum cytokines and chemokines involved in the regulation of vascular function and inflammation are increased.
Collapse
Affiliation(s)
- Letícia Sabioni
- Instituto Nacional de Cardiologia, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Andrea De Lorenzo
- Instituto Nacional de Cardiologia, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Cristiane Lamas
- Instituto Nacional de Cardiologia, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Fabiana Muccillo
- Instituto Nacional de Cardiologia, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Hugo Caire Castro-Faria-Neto
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Estato
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduardo Tibirica
- Instituto Nacional de Cardiologia, Ministério da Saúde, Rio de Janeiro, Brazil.
| |
Collapse
|
17
|
Raucci F, Saviano A, Casillo GM, Guerra-Rodriguez M, Mansour AA, Piccolo M, Ferraro MG, Panza E, Vellecco V, Irace C, Caso F, Scarpa R, Mascolo N, Alfaifi M, Iqbal AJ, Maione F. IL-17-induced inflammation modulates the mPGES-1/PPAR-γ pathway in monocytes/macrophages. Br J Pharmacol 2021; 179:1857-1873. [PMID: 33595097 DOI: 10.1111/bph.15413] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/15/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Recent biochemical and pharmacological studies have reported that in several tissues and cell types, microsomal PGE2 synthase (mPGES) and PPAR-γ expression are modulated by a variety of inflammatory factors and stimuli. Considering that very little is known about the biological effects promoted by IL-17 in the context of mPGES-1/PPAR-γ modulation, we sought to investigate the contribution of this unique cytokine on this integrated pathway during the onset of inflammation. EXPERIMENTAL APPROACH We evaluated effects of PF 9184 (mPGES-1 inhibitor) and troglitazone (PPAR-γ agonist) in vitro, using the mouse macrophage cell line J774A.1. In vivo, the dorsal air pouch model in CD1 mice was used, and inflammatory infiltrates were analysed by flow cytometry. Locally produced cyto-chemokines and PGs were assessed using elisa assays. Western blots were also employed to determine the activity of various enzymes involved in downstream signalling pathways. KEY RESULTS PF 9184 and troglitazone, in a time- and dose-dependent manner, modulated leukocyte infiltration, myeloperoxidase activity, and the expression of COX-2/mPGES-1, NF-кB/IкB-α, and mPTGDS-1/PPAR-γ, induced by IL-17. Moreover, both PF 9184 and troglitazone modulated PG (PGE2 , PGD2 , and PGJ2 ) production, the expression of different pro-inflammatory cyto-chemokines, and the recruitment of inflammatory monocytes, in response to IL-17. CONCLUSIONS AND IMPLICATIONS Our data suggest that IL-17 may constitute a specific modulator of inflammatory monocytes during later phases of the inflammatory response. The results of this study show, for the first time, that the IL-17/mPGES-1/PPAR-γ pathway could represent a potential therapeutic target for inflammatory-based and immune-mediated diseases.
Collapse
Affiliation(s)
- Federica Raucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Anella Saviano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Miguel Guerra-Rodriguez
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Marialuisa Piccolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Grazia Ferraro
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Elisabetta Panza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carlo Irace
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Raffaele Scarpa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Nicola Mascolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Mohammed Alfaifi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Asif Jilani Iqbal
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.,Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
18
|
Scarpa R, Caso F, Costa L, Passavanti S, Vitale MG, Trojaniello C, Del Puente A, Ascierto PA. May the analysis of 1918 influenza pandemic give hints to imagine the possible magnitude of Corona Virus Disease-2019 (COVID-19)? J Transl Med 2020; 18:489. [PMID: 33353549 PMCID: PMC7753514 DOI: 10.1186/s12967-020-02673-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/10/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND In 1918 an unknown infectious agent spread around the world infecting over one-third of the general population and killing almost 50 million people. Many countries were at war, the First World War. Since Spain was a neutral country and Spanish press could report about the infection without censorship, this condition is commonly remembered as "Spanish influenza". This review examines several aspects during the 1918 influenza pandemic to bring out evidences which might be useful to imagine the possible magnitude of the present coronavirus disease 2019 (COVID-19). METHODS In the first part of this review we will examine the origin of the SARS-Coronavirus-2 and 1918 Spanish Influenza Virus and the role played by host and environment in its diffusion. We will also include in our analysis an evaluation of different approaches utilized to restrain the spread of pandemic and to treat infected patients. In the second part, we will try to imagine the magnitude of the present COVID-19 pandemic and the possible measures able to restrain in the present environment its spread. RESULTS Several factors characterize the outcome in a viral pandemic infection. They include the complete knowledge of the virus, the complete knowledge of the host and of the environment where the host lives and the pandemic develops. CONCLUSION By comparing the situation seen in 1918 with the current one, we are now in a more favourable position. The experience of the past teaches us that their success is linked to a rapid, constant and lasting application. Then, rather than coercion, awareness of the need to observe such prevention measures works better.
Collapse
Affiliation(s)
- Raffaele Scarpa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio, Pansini 5, 80131, Naples, Italy.
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio, Pansini 5, 80131, Naples, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio, Pansini 5, 80131, Naples, Italy
| | - Saverio Passavanti
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio, Pansini 5, 80131, Naples, Italy
| | - Maria Grazia Vitale
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Claudia Trojaniello
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Antonio Del Puente
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio, Pansini 5, 80131, Naples, Italy
| | - Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| |
Collapse
|
19
|
Raucci F, Mansour AA, Casillo GM, Saviano A, Caso F, Scarpa R, Mascolo N, Iqbal AJ, Maione F. Interleukin-17A (IL-17A), a key molecule of innate and adaptive immunity, and its potential involvement in COVID-19-related thrombotic and vascular mechanisms. Autoimmun Rev 2020; 19:102572. [PMID: 32376393 PMCID: PMC7252120 DOI: 10.1016/j.autrev.2020.102572] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 04/25/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Guraiger, Abha 62529, Saudi Arabia
| | - Gian Marco Casillo
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Francesco Caso
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, via S. Pansini 5, 80131 Naples, Italy
| | - Raffaele Scarpa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, via S. Pansini 5, 80131 Naples, Italy
| | - Nicola Mascolo
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| |
Collapse
|
20
|
Najem MY, Couturaud F, Lemarié CA. Cytokine and chemokine regulation of venous thromboembolism. J Thromb Haemost 2020; 18:1009-1019. [PMID: 32020753 DOI: 10.1111/jth.14759] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/15/2020] [Accepted: 02/03/2020] [Indexed: 12/27/2022]
Abstract
Morbidity and mortality from venous thromboembolism (VTE), which refers to deep vein thrombosis and pulmonary embolism, have a substantial effect on the global burden of disease. The field of venous thrombosis research has been dramatically changed over the past 10 years with the improvement of animal models that shed some light on the interaction between inflammation and thrombosis. Important recent advances provided evidence of the implication of the innate immune system in venous thrombosis. In this review, we highlighted the cytokines and chemokines that regulate mechanisms of thrombus formation and resolution. Cytokines are pleiotropic, redundant, and multifunctional endogenous mediators orchestrating the inflammatory responses leading to thrombus formation or resolution. The use of experimental models has revealed the pro-thrombotic activity of some cytokines including interferon-γ, interleukin (IL)-6, chemokine ligand 2, IL-17A, IL-9, IL-1β, and transforming growth factor-β. Other cytokines such as IL-10, tumor necrosis factor-α, and IL-8 appear to promote thrombus resolution in late phase of venous thromboembolism. The purpose of this review is to bring together the current knowledge regarding the cytokines and chemokines that have been involved in thrombosis formation and resolution. We postulate that an imbalance between pro-thrombotic and anti-thrombotic cytokines/chemokines may be involved in the pathophysiology of VTE. However, in-depth basic and clinical research in venous thrombosis is still require to fully understand the precise mechanism of action of these cytokines.
Collapse
Affiliation(s)
- Maria Y Najem
- EA3878 (GETBO), Brest Hospital, Univ Brest, Brest, France
| | | | - Catherine A Lemarié
- EA3878 (GETBO), Brest Hospital, Univ Brest, Brest, France
- INSERM 1078, Brest, France
| |
Collapse
|
21
|
CD4 + T cell phenotypes in the pathogenesis of immune thrombocytopenia. Cell Immunol 2020; 351:104096. [PMID: 32199587 DOI: 10.1016/j.cellimm.2020.104096] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/03/2020] [Accepted: 03/12/2020] [Indexed: 12/14/2022]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by low platelet counts due to enhanced platelet clearance and compromised production. Traditionally, ITP was regarded a B cell mediated disorder as anti-platelet antibodies are detected in most patients. The very nature of self-antigens, evident processes of isotype switching and the affinity maturation of anti-platelet antibodies indicate that B cells in order to mount anti-platelet immune response require assistance of auto-reactive CD4+ T cells. For a long time, ITP pathogenesis has been exclusively reviewed through the prism of the disturbed balance between Th1 and Th2 subsets of CD4+ T cells, however, more recently new subsets of these cells have been described including Th17, Th9, Th22, T follicular helper and regulatory T cells. In this paper, we review the current understanding of the role and immunological mechanisms by which CD4+ T cells contribute to the pathogenesis of ITP.
Collapse
|
22
|
Adams B, Nunes JM, Page MJ, Roberts T, Carr J, Nell TA, Kell DB, Pretorius E. Parkinson's Disease: A Systemic Inflammatory Disease Accompanied by Bacterial Inflammagens. Front Aging Neurosci 2019; 11:210. [PMID: 31507404 PMCID: PMC6718721 DOI: 10.3389/fnagi.2019.00210] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/26/2019] [Indexed: 01/08/2023] Open
Abstract
Parkinson’s disease (PD) is a well-known neurodegenerative disease with a strong association established with systemic inflammation. Recently, the role of the gingipain protease group from Porphyromonas gingivalis was implicated in Alzheimer’s disease and here we present evidence, using a fluorescent antibody to detect gingipain R1 (RgpA), of its presence in a PD population. To further elucidate the action of this gingipain, as well as the action of the lipopolysaccharide (LPS) from P. gingivalis, low concentrations of recombinant RgpA and LPS were added to purified fluorescent fibrinogen. We also substantiate previous findings regarding PD by emphasizing the presence of systemic inflammation via multiplex cytokine analysis, and demonstrate hypercoagulation using thromboelastography (TEG), confocal and electron microscopy. Biomarker analysis confirmed significantly increased levels of circulating proinflammatory cytokines. In our PD and control blood analysis, our results show increased hypercoagulation, the presence of amyloid formation in plasma, and profound ultrastructural changes to platelets. Our laboratory analysis of purified fibrinogen with added RgpA, and/or LPS, showed preliminary data with regards to the actions of the protease and the bacterial membrane inflammagen on plasma proteins, to better understand the nature of established PD.
Collapse
Affiliation(s)
- Büin Adams
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - J Massimo Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Martin J Page
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Timothy Roberts
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,Department of Biochemistry, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Jonathan Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Theo A Nell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,Department of Biochemistry, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
23
|
Herster F, Bittner Z, Codrea MC, Archer NK, Heister M, Löffler MW, Heumos S, Wegner J, Businger R, Schindler M, Stegner D, Schäkel K, Grabbe S, Ghoreschi K, Miller LS, Weber ANR. Platelets Aggregate With Neutrophils and Promote Skin Pathology in Psoriasis. Front Immunol 2019; 10:1867. [PMID: 31474979 PMCID: PMC6706802 DOI: 10.3389/fimmu.2019.01867] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/23/2019] [Indexed: 12/13/2022] Open
Abstract
Psoriasis is a frequent systemic inflammatory autoimmune disease characterized primarily by skin lesions with massive infiltration of leukocytes, but frequently also presents with cardiovascular comorbidities. Especially polymorphonuclear neutrophils (PMNs) abundantly infiltrate psoriatic skin but the cues that prompt PMNs to home to the skin are not well-defined. To identify PMN surface receptors that may explain PMN skin homing in psoriasis patients, we screened 332 surface antigens on primary human blood PMNs from healthy donors and psoriasis patients. We identified platelet surface antigens as a defining feature of psoriasis PMNs, due to a significantly increased aggregation of neutrophils and platelets in the blood of psoriasis patients. Similarly, in the imiquimod-induced experimental in vivo mouse model of psoriasis, disease induction promoted PMN-platelet aggregate formation. In psoriasis patients, disease incidence directly correlated with blood platelet counts and platelets were detected in direct contact with PMNs in psoriatic but not healthy skin. Importantly, depletion of circulating platelets in mice in vivo ameliorated disease severity significantly, indicating that both PMNs and platelets may be relevant for psoriasis pathology and disease severity.
Collapse
Affiliation(s)
| | - Zsofia Bittner
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | | | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Martin Heister
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Markus W Löffler
- Department of Immunology, University of Tübingen, Tübingen, Germany.,Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany.,Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany
| | - Simon Heumos
- Quantitative Biology Center, University of Tübingen, Tübingen, Germany
| | - Joanna Wegner
- Department of Dermatology, University Hospital Mainz, Mainz, Germany
| | - Ramona Businger
- Division of Molecular Virology, Institute of Virology, Tübingen, Germany
| | - Michael Schindler
- Division of Molecular Virology, Institute of Virology, Tübingen, Germany
| | - David Stegner
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Knut Schäkel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Hospital Mainz, Mainz, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany.,Department of Dermatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | |
Collapse
|
24
|
Beringer A, Miossec P. Systemic effects of IL-17 in inflammatory arthritis. Nat Rev Rheumatol 2019; 15:491-501. [PMID: 31227819 DOI: 10.1038/s41584-019-0243-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2019] [Indexed: 12/20/2022]
Abstract
Inflammatory arthritis occurs in many diseases and is characterized by joint inflammation and damage. However, the inflammatory state in arthritis is commonly associated with systemic manifestations, which are generally linked to a poor prognosis. The pro-inflammatory cytokine IL-17 functions within a complex network of cytokines and contributes to the pathogenesis of various inflammatory diseases. Three IL-17 inhibitors have already been approved for the treatment of psoriasis, psoriatic arthritis, and ankylosing spondylitis. After a brief description of IL-17 and its local effects on joints, this Review focuses on the systemic effects of IL-17 in inflammatory arthritis. Increased circulating concentrations of bioactive IL-17 mediate changes in blood vessels, liver and cardiac and skeletal muscles. The effects of IL-17 on vascular and cardiac cells might contribute to the increased risk of cardiovascular events that occurs in all patients with inflammatory disorders. In the liver, IL-17 contributes to the high circulating concentrations of acute-phase proteins, such as C-reactive protein, and the appearance of liver lesions. In skeletal muscle, IL-17 contributes to muscle contractibility defects and weakness. Thus, targeting IL-17 might have beneficial effects at both local and systemic levels, and could also be proposed for the treatment of a wider range of inflammatory diseases.
Collapse
Affiliation(s)
- Audrey Beringer
- Immunogenomics and Inflammation Research Unit, EA 4130, University of Lyon, Hospices Civils de Lyon, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit, EA 4130, University of Lyon, Hospices Civils de Lyon, Lyon, France.
| |
Collapse
|
25
|
Zhang Q, Wu H, Zhang J, Qi Q, Zhang W, Xia R. Preoperative Immune Response is Associated with Perioperative Transfusion Requirements in Glioma Surgery. J Cancer 2019; 10:3526-3532. [PMID: 31293657 PMCID: PMC6603405 DOI: 10.7150/jca.28953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 05/05/2019] [Indexed: 01/08/2023] Open
Abstract
Immunosuppression induced by transfusion causes postoperative adverse events including poor prognosis in cancer, but data on influence of the immune response on blood transfusion requirements during perioperative period are limited. The aim of this study was to investigate whether the preoperative immune response is associated with perioperative blood cell transfusion in a glioma surgical patient population. The authors identified 321 cases of surgery for treatment of glioma. Patient variables, preoperative laboratory variables (hemoglobin, platelet count, activated partial thromboplastin time, prothrombin time, hematocrit, red and white blood cell count), and transfusions were registered. Plasma concentration of Th-associated cytokines was measured by flow cytometry. Multivariable regression analysis and receiver operating characteristic curve were undertaken to identify predictors of transfusion. Of 321 patients, 157 (48.90%) received red blood cells transfusion. The mean age is significantly higher in transfusion group compared to no transfusion group, while postoperative hospital stay, preoperative hemoglobin, prothrombin time, activated partial thromboplastin time, platelet count, red and white blood cell count and hematocrit of patients did not differ significantly between the two groups. No significant differences of IL-2, -4, -6, -10 and INF-γ concentration were observed between transfusion and no transfusion group. The concentration of TNF and IL-17A was significantly lower in transfusion patients than in the no transfusion subjects. Low plasma TNF and IL-17A levels predicted high perioperative transfusion rate, the combination of them enlarged the prognostic accuracy of testing. Our study demonstrates that the preoperative immune response influences transfusion requirements, and TNF and IL-17 are important predictive risk factors for perioperative use of blood components in glioma patients.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Huahui Wu
- Harbin Hospital of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Jingjun Zhang
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Qi
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Medical Imaging, Renji Hospital, Medical School of Jiaotong University, Shanghai, China
| | - Rong Xia
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Evidence for the important role of inflammation in xenotransplantation. JOURNAL OF INFLAMMATION-LONDON 2019; 16:10. [PMID: 31148951 PMCID: PMC6537172 DOI: 10.1186/s12950-019-0213-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
There is increasing evidence of a sustained state of systemic inflammation after pig-to-nonhuman primate (NHP) xenotransplantation (that has been termed systemic inflammation in xenograft recipients [SIXR]). Increases in inflammatory markers, e.g., C-reactive protein, histones, serum amyloid A, D-dimer, cytokines, chemokines, and a decrease in free triiodothyronine, have been demonstrated in the recipient NHPs. The complex interactions between inflammation, coagulation, and the immune response are well-recognized, but the role of inflammation in xenograft recipients is not fully understood. The evidence suggests that inflammation can promote the activation of coagulation and the adaptive immune response, but the exact mechanisms remain uncertain. If prolonged xenograft survival is to be achieved, anti-inflammatory strategies (e.g., the administration of anti-inflammatory agents, and/or the generation of genetically-engineered organ-source pigs that are protected from the effect of inflammation) may be necessary to prevent, control, or negate the effect of the systemic inflammation that develops in xenograft recipients. This may allow for a reduction in the intensity of exogenous immunosuppressive therapy. If immunological tolerance to a xenograft is to be obtained, then control of inflammation may be essential.
Collapse
|
27
|
Li Y, Shen Y, Lin D, Zhang H, Wang T, Liu H, Wang Y. Neutrophils and IL17A mediate flagellar hook protein FlgE-induced mouse acute lung inflammation. Cell Microbiol 2018; 21:e12975. [PMID: 30412932 DOI: 10.1111/cmi.12975] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Yuanyuan Li
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, College of Medicine; Soochow University; Suzhou China
| | - Ying Shen
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, College of Medicine; Soochow University; Suzhou China
| | - Dandan Lin
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, College of Medicine; Soochow University; Suzhou China
| | - Hongbo Zhang
- Department of Clinical Laboratory; Qilu Hospital of Shandong University (Qingdao); Qingdao China
| | - Ting Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, College of Medicine; Soochow University; Suzhou China
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology; National University of Singapore; Singapore Singapore
| | - Yiqiang Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, College of Medicine; Soochow University; Suzhou China
| |
Collapse
|
28
|
Maione F, Iqbal AJ, Raucci F, Letek M, Bauer M, D'Acquisto F. Repetitive Exposure of IL-17 Into the Murine Air Pouch Favors the Recruitment of Inflammatory Monocytes and the Release of IL-16 and TREM-1 in the Inflammatory Fluids. Front Immunol 2018; 9:2752. [PMID: 30555461 PMCID: PMC6284009 DOI: 10.3389/fimmu.2018.02752] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/08/2018] [Indexed: 12/21/2022] Open
Abstract
The infiltration of Th17 cells in tissues and organs during the development of many autoimmune diseases is considered a key step toward the establishment of chronic inflammation. Indeed, the localized and prolonged release of IL-17 in specific tissues has been associated with an increased severity of the inflammatory response that remains sustained over time. The cellular and molecular mechanisms behind these effects are far from being clear. In this study we investigated the effects of two repetitive administration of recombinant IL-17 into the murine air pouch to simulate a scenario where IL-17 is released over time in a pre-inflamed tissue. Consistent with our previous observations, mice receiving a single dose of IL-17 showed a transitory influx of neutrophils into the air pouch that peaked at 24 h and declined at 48 h. Conversely, mice receiving a double dose of the cytokine—one at time 0 and the second after 24 h—showed a more dramatic inflammatory response with almost 2-fold increase in the number of infiltrated leukocytes and significant higher levels of TNF-α and IL-6 in the inflammatory fluids. Further analysis of the exacerbated inflammatory response of double-injected IL-17 mice showed a unique cellular and biochemical profile with inflammatory monocytes as the second main population emigrating to the pouch and IL-16 and TREM-1 as the most upregulated cytokines found in the inflammatory fluids. Most interestingly, mice receiving a double injection of IL-1β did not show any change in the cellular or biochemical inflammatory response compared to those receiving a single injection or just vehicle. Collectively these results shed some light on the function of IL-17 as pro-inflammatory cytokine and provide possible novel ways to target therapeutically the pathogenic effects of IL-17 in autoimmune conditions.
Collapse
Affiliation(s)
- Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.,William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Federica Raucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Michal Letek
- Health Science Research Centre, Department of Life Science, University of Roehampton, London, United Kingdom
| | - Martina Bauer
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Fulvio D'Acquisto
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Health Science Research Centre, Department of Life Science, University of Roehampton, London, United Kingdom
| |
Collapse
|
29
|
IL-17A promotes the formation of deep vein thrombosis in a mouse model. Int Immunopharmacol 2018; 57:132-138. [PMID: 29482157 DOI: 10.1016/j.intimp.2018.02.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 02/10/2018] [Accepted: 02/12/2018] [Indexed: 02/07/2023]
Abstract
Deep venous thrombosis (DVT) is a significant problem in the health care industry worldwide. However, the factors and signaling pathways that trigger DVT formation are still largely unknown. In this study, we investigated the role of interleukin-17A (IL-17A) in DVT formation, focusing on the role of platelet aggregation, neutrophil infiltration, and endothelium cell (EC) activation. Notably, IL-17A levels increased in DVT patients as well as in a mouse DVT model. The DVT model mice were injected with recombinant mouse-IL-17A (rIL-17A) or anti-IL-17A monoclonal antibody (mAb) to further evaluate the effects of this cytokine. We found that rIL-17A promotes DVT formation, while IL-17A mAb represses DVT formation. Furthermore, platelet activation, highlighted by CD61 and CD49β expression, and aggregation were enhanced in platelets of rIL-17A-treated mice. rIL-17A also enhanced neutrophil infiltration by regulating the expression of macrophage inflammatory protein-2 (MIP-2) and the release of neutrophil extracellular traps (NETs). IL-17A mAb treatment inhibited both platelet activation and neutrophil activity. Moreover, rIL-17A appears to promote vein EC activation, while IL-17A mAb deters it. Taken together, these data suggest that IL-17A promotes DVT pathogenesis by enhancing platelet activation and aggregation, neutrophil infiltration, and EC activation and that anti-IL-17A mAb could be used for the treatment of DVT.
Collapse
|
30
|
|
31
|
Lawrence SM, Ruoss JL, Wynn JL. IL-17 in neonatal health and disease. Am J Reprod Immunol 2017; 79:e12800. [PMID: 29243317 DOI: 10.1111/aji.12800] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 11/21/2017] [Indexed: 12/11/2022] Open
Abstract
Over the last few years, scientific interest in the cytokine IL-17A has intensified as its role in human health and disease has been elucidated. Discovered almost a quarter century ago, IL-17A is known to have poor biologic activity when acting alone, but attains robust actions when working synergistically with potent mediators of proinflammatory immune responses, such as IL-6 and IL-8. IL-17A is produced by specialized innate immune cells that protect host barriers from the outside world. Like sentries, these innate immune cells can "sound the alarm" through increased production of IL-17A, causing activation and recruitment of primed neutrophils and monocytes when pathogens escape initial host defenses. In this way, IL-17A promulgates mechanisms responsible for pathogen death and clearance. However, when IL-17A pathways are triggered during fetal development, due to chorioamnionitis or in utero inflammatory conditions, IL-17A can instigate and/or exacerbate fetal inflammatory responses that increase neonatal morbidities and mortality associated with common neonatal conditions such as sepsis, bronchopulmonary dysplasia (BPD), patent ductus arteriosus (PDA), and necrotizing enterocolitis (NEC). This review details the ontogeny of IL-17A in the fetus and newborn, discusses how derangements in its production can lead to pathology, and describes known and evolving therapies that may attenuate IL-17A-mediated human conditions.
Collapse
Affiliation(s)
- Shelley M Lawrence
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, College of Medicine, University of California, San Diego, La Jolla, CA, USA.,Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Jessica Lauren Ruoss
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - James L Wynn
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA.,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
32
|
Li X, Teeling P, Mackaay C, Ploegmakers H, van der Loos C, Daemen M, de Winter R, van der Wal A, de Boer O. Neutrophils, neutrophil extracellular traps and interleukin-17 associate with the organisation of thrombi in acute myocardial infarction. Thromb Haemost 2017; 109:290-7. [DOI: 10.1160/th12-06-0425] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 11/08/2012] [Indexed: 01/09/2023]
Abstract
SummaryNeutrophils are important cellular sources of interleukin (IL) 17A and –F. Moreover, upon activation neutrophils are able to excrete chromatin embedded with components from their cytoplasmic granules to form ‘neutrophil extracellular traps’ (NETs). Recent studies suggested that NETs contribute to thrombosis by promoting fibrin deposition and platelet aggregation. IL17A may also promote thrombosis by enhancing platelet aggregation. In the present study we investigated the presence of neutrophils, NETs and IL17A and –F in coronary thrombosuction specimens obtained from patients after acute myocardial infarction. Neutrophils and NETs were identified using histochemical (H&E, Feulgen procedure) and immunohistochemical stainings (Histone H1, myeloperoxidase, neutrophil elastase) in 15 fresh, 15 lytic and 15 organised thrombi. The presence and distribution of IL17A and –F was studied using (immuno)histochemical double staining and spectral image analysis, rtPCR and Western blot. High numbers of neutrophils are present (10–30% of the thrombus mass) in fresh and lytic, but not in organized thrombus. NETs were frequently observed in fresh (4/15) and lytic (12/15), but never in organised thrombus specimens. Double staining combining the Feulgen reaction with Histone- H1, MPO or neutrophil elastase confirmed colocalisation with DNA. Cytoplasmatic IL17A/F staining was found in the majority of the neutrophils, extracellularly and in NETs. Western blotting confirmed the presence of IL17A and IL17F in thrombus specimens. In conclusion, a large burden of neutrophils, neutrophil extracellular traps and IL17A and –F are important constituents of fresh and lytic thrombus after acute myocardial infarction. The specific colocalisation of these indicates a role during thrombus stabilisation and growth.
Collapse
|
33
|
Li Y, Golden JB, Camhi MI, Zhang X, Fritz Y, Diaconu D, Ivanco TL, Simon DI, Kikly K, McCormick TS, Wang Y, Ward NL. Protection from Psoriasis-Related Thrombosis after Inhibition of IL-23 or IL-17A. J Invest Dermatol 2017; 138:310-315. [PMID: 28951241 DOI: 10.1016/j.jid.2017.09.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/29/2017] [Accepted: 09/12/2017] [Indexed: 12/19/2022]
Abstract
Psoriasis patients experience chronic systemic skin inflammation and develop cardiovascular comorbidities that shorten their lifespan. Whether cardiovascular disease is improved by treatment with current biologics that target disease-specific pathways is unclear. KC-Tie2 mice develop psoriasiform skin inflammation with increases in IL-23 and IL-17A and proinflammatory monocytosis and neutrophilia that precedes development of carotid artery thrombus formation. To examine whether targeted blockade of IL-23 or IL-17A in KC-Tie2 psoriasis mice improves cardiovascular outcomes, mice were treated systemically for 6 weeks with antibodies targeting IL-17A, IL-17RA, IL-12/23p40, or IL-23p19. Skin inflammation; thrombosis clotting times; and percentage of splenic monocytes, neutrophils, and CD4 T cells were examined. Skin inflammation significantly improved in KC-Tie2 mice treated with each of the antibodies targeting IL-23, IL-17A, or IL-17RA, consistent with clinical efficacy observed in psoriasis patients. The time to occlusive thrombus formation lengthened in these mice and correlated with attenuated acanthosis. This decrease in skin inflammation paralleled decreases in splenic neutrophils (CD11b+Ly6G+) but not monocytes (CD11b+Ly6Chigh) or T cells (CD4+). Our data show that targeted inhibition of IL-23 or IL-17A improves psoriasis-like skin disease and also improves cardiovascular disease in mice.
Collapse
Affiliation(s)
- Yumeng Li
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jackelyn B Golden
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Maya I Camhi
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Xiufen Zhang
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yi Fritz
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Doina Diaconu
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Tammy L Ivanco
- Department of Psychology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Daniel I Simon
- Department of Medicine, Division of Cardiology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA; Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Thomas S McCormick
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio, USA; Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Yunmei Wang
- Department of Medicine, Division of Cardiology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA; Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nicole L Ward
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio, USA; Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.
| |
Collapse
|
34
|
Robert M, Miossec P. Effects of Interleukin 17 on the cardiovascular system. Autoimmun Rev 2017; 16:984-991. [PMID: 28705781 DOI: 10.1016/j.autrev.2017.07.009] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 06/17/2017] [Indexed: 12/25/2022]
Abstract
Cardiovascular diseases remain the leading cause of death worldwide and account for most of the premature mortality observed in chronic inflammatory diseases. Common mechanisms underlie these two types of disorders, where the contribution of Interleukin (IL)-17A, the founding member of the IL-17 family, is highly suspected. While the local effects of IL-17A in inflammatory disorders have been well described, those on the cardiovascular system remain less studied. This review focuses on the effects of IL-17 on the cardiovascular system both on isolated cells and in vivo. IL-17A acts on vessel and cardiac cells, leading to inflammation, coagulation and thrombosis. In vivo and clinical studies have shown its involvement in the pathogenesis of cardiovascular diseases including atherosclerosis and myocardial infarction that occur prematurely in chronic inflammatory disorders. As new therapeutic approaches are targeting the IL-17 pathway, this review should help to better understand their positive and negative outcomes on the cardio-vascular system.
Collapse
Affiliation(s)
- Marie Robert
- Immunogenomics and Inflammation Research Unit, EA 4130, Department of Immunology and Rheumatology, University of Lyon, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit, EA 4130, Department of Immunology and Rheumatology, University of Lyon, Lyon, France.
| |
Collapse
|
35
|
Pietronigro EC, Della Bianca V, Zenaro E, Constantin G. NETosis in Alzheimer's Disease. Front Immunol 2017; 8:211. [PMID: 28303140 PMCID: PMC5332471 DOI: 10.3389/fimmu.2017.00211] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 02/15/2017] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by the progressive deterioration of cognitive functions. Its neuropathological features include amyloid-β (Aβ) accumulation, the formation of neurofibrillary tangles, and the loss of neurons and synapses. Neuroinflammation is a well-established feature of AD pathogenesis, and a better understanding of its mechanisms could facilitate the development of new therapeutic approaches. Recent studies in transgenic mouse models of AD have shown that neutrophils adhere to blood vessels and migrate inside the parenchyma. Moreover, studies in human AD subjects have also shown that neutrophils adhere and spread inside brain vessels and invade the parenchyma, suggesting these cells play a role in AD pathogenesis. Indeed, neutrophil depletion and the therapeutic inhibition of neutrophil trafficking, achieved by blocking LFA-1 integrin in AD mouse models, significantly reduced memory loss and the neuropathological features of AD. We observed that neutrophils release neutrophil extracellular traps (NETs) inside blood vessels and in the parenchyma of AD mice, potentially harming the blood–brain barrier and neural cells. Furthermore, confocal microscopy confirmed the presence of NETs inside the cortical vessels and parenchyma of subjects with AD, providing more evidence that neutrophils and NETs play a role in AD-related tissue destruction. The discovery of NETs inside the AD brain suggests that these formations may exacerbate neuro-inflammatory processes, promoting vascular and parenchymal damage during AD. The inhibition of NET formation has achieved therapeutic benefits in several models of chronic inflammatory diseases, including autoimmune diseases affecting the brain. Therefore, the targeting of NETs may delay AD pathogenesis and offer a novel approach for the treatment of this increasingly prevalent disease.
Collapse
Affiliation(s)
| | - Vittorina Della Bianca
- Department of Medicine, Section of General Pathology, University of Verona , Verona , Italy
| | - Elena Zenaro
- Department of Medicine, Section of General Pathology, University of Verona , Verona , Italy
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona , Verona , Italy
| |
Collapse
|
36
|
Essential roles for platelets during neutrophil-dependent or lymphocyte-mediated defense against bacterial pathogens. Blood Coagul Fibrinolysis 2017; 27:667-72. [PMID: 26588444 DOI: 10.1097/mbc.0000000000000455] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Emerging evidence from animal models suggests that platelets may participate in a wide variety of processes including the immune response against infection. More than 200 whole blood samples from patients and healthy controls were run in the System XE-5000 analyzer, and plasma fractions were separated for the following tests by ELISA, Luminex and light scattering. We describe two mechanisms by which platelets may contribute to immune function against various bacterial pathogens based on increased mean platelet volume in gram-positive bacterial infections and increased platelet counts in gram-negative bacterial infections. Gram-negative bacteria activate platelets to recruit neutrophils, which participate in the immune response against infection. During this process, fractalkine, macrophage inflammatory protein-1β, interleukin-17A, tumor necrosis factor-α and platelet-activating factor were higher in patients infected with Escherichia coli; additionally, giant platelets were observed under the microscope. Meanwhile, we found that platelets played a different role in gram-positive bacterial infections. Specifically, they could actively adhere to gram-positive bacteria in circulation and transfer them to immune sites to promote antibacterial lymphocyte expansion. During this process, complement C3 and factor XI were more highly expressed in patients infected with Staphylococcus aureus; additionally, we detected more small platelets under the microscope. Platelets participate in the immune response against both gram-negative and gram-positive bacteria, although the mechanisms differ. These results will help us understand the complex roles of platelets during infections, and direct our use of antibiotics based on clinical platelet data.
Collapse
|
37
|
Ottaiano TF, Andrade SS, de Oliveira C, Silva MCC, Buri MV, Juliano MA, Girão MJBC, Sampaio MU, Schmaier AH, Wlodawer A, Maffei FHA, Oliva MLV. Plasma kallikrein enhances platelet aggregation response by subthreshold doses of ADP. Biochimie 2017; 135:72-81. [PMID: 28115185 DOI: 10.1016/j.biochi.2017.01.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/16/2016] [Accepted: 01/18/2017] [Indexed: 11/26/2022]
Abstract
Human plasma kallikrein (huPK) potentiates platelet responses to subthreshold doses of ADP, although huPK itself, does not induce platelet aggregation. In the present investigation, we observe that huPK pretreatment of platelets potentiates ADP-induced platelet activation by prior proteolysis of the G-protein-coupled receptor PAR-1. The potentiation of ADP-induced platelet activation by huPK is mediated by the integrin αIIbβ3 through interactions with the KGD/KGE sequence motif in huPK. Integrin αIIbβ3 is a cofactor for huPK binding to platelets to support PAR-1 hydrolysis that contributes to activation of the ADP signaling pathway. This activation pathway leads to phosphorylation of Src, AktS473, ERK1/2, and p38 MAPK, and to Ca2+ release. The effect of huPK is blocked by specific antagonists of PAR-1 (SCH 19197) and αIIbβ3 (abciximab) and by synthetic peptides comprising the KGD and KGE sequence motifs of huPK. Further, recombinant plasma kallikrein inhibitor, rBbKI, also blocks this entire mechanism. These results suggest a new function for huPK. Formation of plasma kallikrein lowers the threshold for ADP-induced platelet activation. The present observations are consistent with the notion that plasma kallikrein promotes vascular disease and thrombosis in the intravascular compartment and its inhibition may ameliorate cardiovascular disease and thrombosis.
Collapse
Affiliation(s)
- Tatiana F Ottaiano
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | - Sheila S Andrade
- Department of Gynecology, Universidade Federal de São Paulo, São Paulo 04024-002, Brazil; Charitable Association of Blood Collection - COLSAN São Paulo, SP, Brazil
| | - Cleide de Oliveira
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | - Mariana C C Silva
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | - Marcus V Buri
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | - Maria A Juliano
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | - Manoel J B C Girão
- Department of Gynecology, Universidade Federal de São Paulo, São Paulo 04024-002, Brazil; Charitable Association of Blood Collection - COLSAN São Paulo, SP, Brazil
| | - Misako U Sampaio
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | - Alvin H Schmaier
- Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Alexander Wlodawer
- Macromolecular Crystallography Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Francisco H A Maffei
- Department of Orthopedics and Surgery, Universidade Estadual Paulista, Botucatu, Brazil
| | - Maria Luiza V Oliva
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil.
| |
Collapse
|
38
|
Maione F. Commentary: IL-17 in Chronic Inflammation: From Discovery to Targeting. Front Pharmacol 2016; 7:250. [PMID: 27561214 PMCID: PMC4980561 DOI: 10.3389/fphar.2016.00250] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 07/28/2016] [Indexed: 11/14/2022] Open
Affiliation(s)
- Francesco Maione
- Department of Pharmacy, University of Naples Federico II Naples, Italy
| |
Collapse
|
39
|
IL-17 Induces MPTP opening through ERK2 and P53 signaling pathway in human platelets. ACTA ACUST UNITED AC 2015; 35:679-683. [PMID: 26489621 DOI: 10.1007/s11596-015-1489-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 09/09/2015] [Indexed: 02/02/2023]
Abstract
The opening of mitochondrial permeability transition pore (MPTP) plays a critical role in platelet activation. However, the potential trigger of the MPTP opening in platelet activation remains unknown. Inflammation is the crucial trigger of platelet activation. In this study, we aimed to explore whether and how the important inflammatory cytokine IL-17 is associated with MPTP opening in platelets activation by using MPTP inhibitor cyclosporine-A (CsA). The mitochondrial membrane potential (ΔΨm) was detected to reflect MPTP opening levels. And the platelet aggregation, activation, and the primary signaling pathway were also tested. The results showed that the MPTP opening levels were increased and Δψm reduced in platelets administrated with IL-17. Moreover, the levels of aggregation, CD62P, PAC-1, P53 and the phosphorylation of ERK2 were enhanced along with the MPTP opening in platelets pre-stimulated with IL-17. However, CsA attenuated these effects triggered by IL-17. It was suggested that IL-17 could induce MPTP opening through ERK2 and P53 signaling pathway in platelet activation and aggregation.
Collapse
|
40
|
Ro JY, Ryu JH, Park HJ, Cho HJ. Onion (Allium cepa L.) peel extract has anti-platelet effects in rat platelets. SPRINGERPLUS 2015; 4:17. [PMID: 25628983 PMCID: PMC4303602 DOI: 10.1186/s40064-015-0786-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 01/02/2015] [Indexed: 11/10/2022]
Abstract
The effects of onion peel extract (OPE) in collagen (5 μg/mL)-stimulated washed rat platelet aggregation were investigated. OPE inhibited platelet aggregation via inhibition of aggregation-inducing molecules, intracellular Ca(2+) and thromboxane A2 (TXA2) by blocking cyclooxygenase-1 (COX-1) and TXA2 synthase (TXAS) activities in a dose-dependent manner. In addition, OPE elevated the formation of cyclic adenosine monophosphate (cAMP), aggregation-inhibiting molecule, but not cyclic guanosine monophosphate (cGMP). High performance liquid chromatography (HPLC) analysis of OPE revealed that OPE contains quercetin, one of the major flavonoids, which has anti-platelet effect. In conclusion, we suggest that OPE is an effective inhibitor of collagen-stimulated platelet aggregation in vitro. Therefore, it can be a promising and safe strategy for anti-cardiovascular diseases.
Collapse
Affiliation(s)
- Ju-Ye Ro
- Department of Biomedical Laboratory Science, College of Medical Science, Konyang University, 685, Gasuwon-dong, Seo-gu, Daejeon 302-718 Korea
| | - Jin-Hyeob Ryu
- Department of Biomedical Laboratory Science, College of Medical Science, Konyang University, 685, Gasuwon-dong, Seo-gu, Daejeon 302-718 Korea.,Present address: Department of Microbiology and Immunology, Institute of Medical Science, Tokyo University, Minato-ku, Tokyo, 108-8639 Japan
| | - Hwa-Jin Park
- Department of Biomedical Laboratory Science, College of Biomedical Science and Engineering and Regional Research Center, Inje University, 607, Obang-dong, Gimhae, Gyungnam 621-749 Korea
| | - Hyun-Jeong Cho
- Department of Biomedical Laboratory Science, College of Medical Science, Konyang University, 685, Gasuwon-dong, Seo-gu, Daejeon 302-718 Korea
| |
Collapse
|
41
|
Reduced IL-35 levels are associated with increased platelet aggregation and activation in patients with acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Ann Hematol 2014; 94:837-45. [PMID: 25512184 DOI: 10.1007/s00277-014-2278-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 12/07/2014] [Indexed: 01/30/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is a major complication associated with allogeneic hematopoietic stem cell transplantation (allo-HSCT). Interleukin (IL)-35 is a novel anti-inflammatory cytokine that suppresses the immune response. This prospective study explored IL-35 plasma levels in 65 patients after HSCT. The results revealed that the peripheral blood of patients with grades III-IV aGVHD (23.46 ng/ml) had reduced IL-35 compared to transplanted patients with grades I-II aGVHD (40.26 ng/ml, p < 0.01) or patients without aGVHD (41.40 ng/ml, p < 0.05). Allografts, including granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood progenitor cell (PBPC) and G-CSF-primed bone marrow (GBM), from 38 patients were analyzed for IL-35 levels with respect to aGVHD. The patients who received lower levels of IL-35 cells in the GBM (28.0 ng/ml, p = 0.551) or lower levels of IL-35 in PBPC (53.46 ng/ml, p = 0.03) exhibited a higher incidence of aGVHD. Patients with aGVHD have increased platelet aggregation. IL-35 was added to patient blood in vitro, and platelet aggregation was inhibited by IL-35 in a dose-dependent manner. The markers of platelet activation (CD62P/PAC-1) can also be inhibited by IL-35. The results indicate that IL-35 may affect the development of aGVHD by inhibiting platelet activation and aggregation. Our data suggests that IL-35 represents a potentially effective therapeutic agent against aGVHD after allo-HSCT.
Collapse
|
42
|
Maione F, De Feo V, Caiazzo E, De Martino L, Cicala C, Mascolo N. Tanshinone IIA, a major component of Salvia milthorriza Bunge, inhibits platelet activation via Erk-2 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2014; 155:1236-42. [PMID: 25038434 DOI: 10.1016/j.jep.2014.07.010] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 06/30/2014] [Accepted: 07/07/2014] [Indexed: 05/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The roots of Salvia milthorriza Bunge (Lamiaceae) known as "Danshen", are used in Traditional Chinese Medicine as a remedy for activating blood and eliminating stasis. TIIA, a diterpenoid of Salvia milthorriza, is one of active components in Danshen that exhibits a significant improvement of the blood flow in the coronary circulatory system and a reduction of myocardial infarction. However, its effect on platelet and underlying mechanism remains largely unknown. On this basis, this compound could be a promising agent to improve blood viscosity and microcirculation and to prevent CVD. MATERIALS AND METHODS In order to investigate the effects of TIIA on platelet functionality and its interaction with various platelet activation pathways, rat PRP were incubated with TIIA for 1 min at 37°C prior the addition of the stimuli (ADP or collagen). Aggregation was monitored in a light transmission aggregometer measuring changes in turbidity with continuous observation up to 10 min after the addition of the stimuli. MAPK signaling pathway and tubulin acetylation were analyzed by a Western blot technique. The effect of the TIIA was also studied in vivo on bleeding time in mice. RESULTS TIIA selectively inhibited rat platelet aggregation induced by reversible ADP stimuli (3 μM) in a concentration-dependent manner (0.5-50 μM). Nevertheless, TIIA was less active against the irreversible stimuli induced by ADP (10 μM) and collagen (10 μg/mL). Moreover, experiments performed on platelet lysates collected at different time-point after the addition of the stimuli shown that TIIA modulated tubulin acetylation and inhibited Erk-2 phosphorylation. Concomitantly, TIIA administrated i.p. at 10 mg/kg significantly amplified the mice bleeding time with an increase of 58% compared to its control (2.06±0.29 min vs 1.30±0.07). ASA was used as reference drug for in vitro and in vivo experiments. CONCLUSIONS This study clarifies the intracellular signaling pathway involved in antiplatelet action of TIIA and also gives preliminary evidences for its anticoagulant activity. On this basis, this compound could be a promising agent to improve blood viscosity and microcirculation and to prevent CVD.
Collapse
Affiliation(s)
- Francesco Maione
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, Via Ponte don Melillo 1, 84084 Fisciano, Salerno, Italy
| | - Elisabetta Caiazzo
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Laura De Martino
- Department of Pharmacy, University of Salerno, Via Ponte don Melillo 1, 84084 Fisciano, Salerno, Italy
| | - Carla Cicala
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Nicola Mascolo
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| |
Collapse
|
43
|
Th17 cells in autoimmune and infectious diseases. Int J Inflam 2014; 2014:651503. [PMID: 25152827 PMCID: PMC4137509 DOI: 10.1155/2014/651503] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 07/20/2014] [Indexed: 02/06/2023] Open
Abstract
The view of CD4 T-cell-mediated immunity as a balance between distinct lineages of Th1 and Th2 cells has changed dramatically. Identification of the IL-17 family of cytokines and of the fact that IL-23 mediates the expansion of IL-17-producing T cells uncovered a new subset of Th cells designated Th17 cells, which have emerged as a third independent T-cell subset that may play an essential role in protection against certain extracellular pathogens. Moreover, Th17 cells have been extensively analyzed because of their strong association with inflammatory disorders and autoimmune diseases. Also, they appear to be critical for controlling these disorders. Similar to Th1 and Th2 cells, Th17 cells require specific cytokines and transcription factors for their differentiation. Th17 cells have been characterized as one of the major pathogenic Th cell populations underlying the development of many autoimmune diseases, and they are enhanced and stabilized by IL-23. The characteristics of Th17 cells, cytokines, and their sources, as well as their role in infectious and autoimmune diseases, are discussed in this review.
Collapse
|
44
|
|
45
|
Interleukin-17A Exacerbates Ferric Chloride-Induced Arterial Thrombosis in Rat Carotid Artery. Int J Inflam 2014; 2014:247503. [PMID: 24940514 PMCID: PMC3997091 DOI: 10.1155/2014/247503] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/11/2014] [Indexed: 02/06/2023] Open
Abstract
Interleukin-17A (IL-17A), the most widely studied member of the IL-17 cytokine family, is a cytokine which emerged to be critical for host defense as well as in the pathogenesis of autoimmune disorders. Moreover, IL-17A is involved in the pathogenesis of cardiovascular diseases, such as atherosclerosis and acute coronary syndrome and in the cardiovascular risk associated with systemic immunological disorders. Consistent with this, we have recently shown that IL-17A increases human and murine platelet response to ADP. In this study we expanded our previous observation and we describe for the first time an in vivo prothrombotic effect of the cytokine. Our results show that IL-17A is synergic with a low FeCl3 concentration in inducing carotid thrombus in rats and suggest that the effect is likely related to a downregulation of CD39 vascular expression and hydrolyzing activity. Our findings indicate that IL-17A might be an important molecule at the interface between hemostasis and inflammation. “This paper is dedicated to the memory of Professor Alfredo Colonna”
Collapse
|
46
|
Su SA, Ma H, Shen L, Xiang MX, Wang JA. Interleukin-17 and acute coronary syndrome. J Zhejiang Univ Sci B 2014; 14:664-9. [PMID: 23897784 DOI: 10.1631/jzus.bqicc701] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Inflammation plays an important role in atherosclerosis, which is also crucial for acute coronary syndrome (ACS). Recent studies have revealed that interleukin (IL)-17, which was regarded as a pro-inflammatory cytokine, has a dual function in the progress of ACS. In this review, we sum up both experimental and clinical studies on the relevance of IL-17 to atherosclerosis and its complications, and summarize the research progress on the effect of IL-17 on the atherosclerotic plaque stability and ACS onset. Although the studies are controversial and the mechanism remains unclear, we highlight the knowledge of the role of IL-17 in ACS and elucidate its potential mechanism.
Collapse
Affiliation(s)
- Sheng-an Su
- Cardiovascular Key Lab of Zhejiang Province, Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | | | | | | | | |
Collapse
|
47
|
Shi G, Field DJ, Ko KA, Ture S, Srivastava K, Levy S, Kowalska MA, Poncz M, Fowell DJ, Morrell CN. Platelet factor 4 limits Th17 differentiation and cardiac allograft rejection. J Clin Invest 2014; 124:543-52. [PMID: 24463452 DOI: 10.1172/jci71858] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 11/07/2013] [Indexed: 01/12/2023] Open
Abstract
Th cells are the major effector cells in transplant rejection and can be divided into Th1, Th2, Th17, and Treg subsets. Th differentiation is controlled by transcription factor expression, which is driven by positive and negative cytokine and chemokine stimuli at the time of T cell activation. Here we discovered that chemokine platelet factor 4 (PF4) is a negative regulator of Th17 differentiation. PF4-deficient and platelet-deficient mice had exaggerated immune responses to cardiac transplantation, including increased numbers of infiltrating Th17 cells and increased plasma IL-17. Although PF4 has been described as a platelet-specific molecule, we found that activated T cells also express PF4. Furthermore, bone marrow transplantation experiments revealed that T cell-derived PF4 contributes to a restriction in Th17 differentiation. Taken together, the results of this study demonstrate that PF4 is a key regulator of Th cell development that is necessary to limit Th17 differentiation. These data likely will impact our understanding of platelet-dependent regulation of T cell development, which is important in many diseases, in addition to transplantation.
Collapse
|
48
|
Hot A, Lavocat F, Lenief V, Miossec P. Simvastatin inhibits the pro-inflammatory and pro-thrombotic effects of IL-17 and TNF-α on endothelial cells. Ann Rheum Dis 2013; 72:754-60. [PMID: 22915623 DOI: 10.1136/annrheumdis-2012-201887] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Statins are widely used for primary and secondary prevention of coronary atherosclerosis. Simvastatin, besides its lipid lowering properties, has various anti-inflammatory effects. The aim of this study was to assess whether simvastatin modulates the vascular effects of interleukin (IL)-17, an emerging actor in atherosclerosis. METHODS The effect of simvastatin was assessed in human umbilical vein endothelial cells treated by IL-17 alone or combined with tumour necrosis factor (TNF)-α, with or without mevalonate, an inhibitor of simvastatin. Its effects on IL-17-induced cytokine or chemokine expression were assessed at the mRNA level using qRT-PCR or protein level by ELISA. Its effect on the IL-17-induced pro-thrombotic state and cell invasion was assessed using a lumi-aggregometer and a Matrigel assay, respectively. RESULTS Simvastatin decreased IL-17-induced IL-6, IL-8, CX3CL-1, RANTES mRNA and CX3CL-1 and CCL20 production. Simvastatin restored the level of IL-33 mRNA which was decreased by IL-17. It reduced the expression of IL-17-induced pro-thrombotic genes such as tissue factor. Simvastatin restored the level of platelet aggregation to normal levels. Simvastatin enhanced the expression of CD39 and thrombomodulin mRNA initially reduced by IL-17 and TNF-α combination. Simvastatin suppressed IL-17-induced endothelial cells invasion. All these effects were reversed by the addition of mevalonate. Finally, simvastatin had an additive effect with infliximab to decrease the effect of the combination of IL-17 and TNF-α on IL-6 mRNA expression. Similar conclusion was obtained with rosuvastatin. CONCLUSIONS Statins inhibit the pro-inflammatory, thrombotic and pro-aggregation effects of IL-17 on vessels. This provides a new understanding of the beneficial effects of statins in blood vessel inflammation.
Collapse
Affiliation(s)
- Arnaud Hot
- Department of Immunology and Rheumatology and the Immunogenomics and inflammation, Hospices Civils de Lyon, University of Lyon 1 mixed research unit, Hospital Edouard Herriot, Lyon, France
| | | | | | | |
Collapse
|
49
|
Zhang S, Yuan J, Yu M, Fan H, Guo ZQ, Yang R, Guo HP, Liao YH, Wang M. IL-17A facilitates platelet function through the ERK2 signaling pathway in patients with acute coronary syndrome. PLoS One 2012; 7:e40641. [PMID: 22808218 PMCID: PMC3394751 DOI: 10.1371/journal.pone.0040641] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 06/11/2012] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Platelet aggregation mediated by inflammation played a critical role in the development of coronary heart diseases (CHD). Our previous clinical researches showed that Th17 cells and their characteristic cytokine IL-17A were associated with the plaque destabilization in patients with acute coronary syndrome (ACS). However, the potent effect of IL-17A on platelets-induced atherothrombosis remains unknown. METHODS AND RESULTS In this study, we detected the plasma IL-17A levels and platelet aggregation in patients with stable angina (SA), unstable angina (UA), acute myocardial infarction (AMI) and chest pain syndrome (CPS). In addition, the markers of platelet activation (CD62P/PAC-1) and the mitogen-activated protein kinases (MAPKs) pathway were detected in platelets from ACS patients. We found that plasma IL-17A levels and platelet aggregation in patients with ACS (UA and AMI) were significantly higher than patients with SA and CPS, and the plasma IL-17A levels were positively correlated with the platelet aggregation (R = 0.47, P<0.01). In addition, in patients with ACS, the platelet aggregation, CD62P/PAC-1 and the phosphorylation of ERK2 signaling pathway were obviously elevated in platelets pre-stimulated with IL-17A in vitro. Furthermore, the specific inhibitor of ERK2 could attenuate platelet aggregation and activation triggered by IL-17A. CONCLUSION Our experiment firstly proved that IL-17A could promote platelet function in patients with ACS via activating platelets ERK2 signaling pathway and may provide a novel target for antiplatelet therapies in CHD.
Collapse
Affiliation(s)
- Shuang Zhang
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yuan
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Miao Yu
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Fan
- Department of Emergency Medicine, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang-Qiang Guo
- Department of Cardiology, Pu Ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Yang
- Department of Hematology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - He-Ping Guo
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Hua Liao
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
50
|
Increased activity of interleukin-23/interleukin-17 cytokine axis in primary antiphospholipid syndrome. Immunobiology 2012; 218:186-91. [PMID: 22559912 DOI: 10.1016/j.imbio.2012.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 03/20/2012] [Indexed: 01/09/2023]
Abstract
The aim of the study was to investigate serum concentrations of interleukin (IL)-17 and IL-17-inducing cytokines IL-23 and transforming growth factor (TGF)-β, as well as IL-17 single nucleotide polymorphism (SNP) rs2275913 in patients with primary antiphospholipid syndrome (PAPS). We studied fifty patients with PAPS and fifty age- and sex-matched healthy controls. The cytokine levels were measured by ELISA, while the rs2275913 SNP located in promoter region of IL-17 gene was genotyped using real-time PCR. The significantly higher levels of IL-17 (p=0.002), IL-23 (p<0.001) and TGF-β (p=0.042) were found in PAPS patients (median 13.1, 9.4, and 125.6 pg/ml, respectively) compared to the control group (6.8, 4.9 and 44.4 pg/ml). There was a significant positive correlation between concentrations of IL-17 and IL-23 (r=0.540, p<0.001), but not between those of IL-17 and TGF-β. No statistically significant differences were observed in the distribution of genotypes and alleles of the IL-17 rs2275913 variants in patients with PAPS compared to healthy subjects. The blood concentrations of IL-17 did not differ in subjects with different rs2275913 genotypes or patients with or without antiphospholipid antibodies. Finally, a trend toward higher IL-17 levels (p=0.063) and the significantly higher IL-17 concentrations (p=0.012) were observed in PAPS patients with deep vein thrombosis and thrombocytopenia, respectively. These data demonstrate that IL-23/IL-17 axis, stimulated independently of TGF-β increase IL-17A gene polymorphism and antiphospholipid antibody production, might contribute to vascular manifestations of PAPS.
Collapse
|