1
|
Yang Z, Wu Y, Huang S, Bao J, Xu L, Fan Y. Risk factors associated with thrombocytopenia in systemic lupus erythematosus: A systematic review and meta-analysis. Autoimmun Rev 2025; 24:103721. [PMID: 39667603 DOI: 10.1016/j.autrev.2024.103721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) frequently manifests with thrombocytopenia (TP), a hematologic complication that heightens the risk of severe outcomes and increases mortality. This meta-analysis aims to evaluate the potential risk factors associated with TP in SLE patients, providing insights into the demographic features, clinical features, and laboratory findings that contribute to this condition. METHODS A comprehensive literature search was conducted across eight databases from inception to September 1, 2024. Study quality was assessed using the Newcastle-Ottawa Scale. Meta-analysis was conducted using univariate and multivariate analyses with Revman 5.3, while heterogeneity was addressed through subgroup and sensitivity analyses. Publication bias was assessed using funnel plots and Egger tests via Stata 15.0. RESULTS Seventeen high-quality studies meeting the inclusion criteria were incorporated into this meta-analysis. Independent risk factors for TP in SLE included age (Demographic Features), serositis, splenomegaly, blood system involvement, and renal involvement (Clinical Features), as well as cardiac involvement, anemia, leukocytopenia, low C3/C4, ACA, and CRP (Laboratory Findings). Arthritis and rash were protective factors. Subgroup analysis addressed heterogeneity caused by unit and sample size differences. Sensitivity analysis comparing the consistency between fixed-effects model (FEM) and random-effects model (REM) confirmed the reliability of the findings, and both funnel plots and Egger tests suggested no publication bias. CONCLUSION This meta-analysis identified several potential independent risk factors for TP in SLE. Early screening and timely intervention for patients with these risk factors are essential to reduce the likelihood of TP, prevent severe organ damage, and improve overall prognosis.
Collapse
Affiliation(s)
- Ze Yang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yanzuo Wu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shuo Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jie Bao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Li Xu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yongsheng Fan
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310005, China.
| |
Collapse
|
2
|
Vostatek R, Ay C. Biological Aging and Venous Thromboembolism: A Review of Telomeres and Beyond. Biomedicines 2024; 13:15. [PMID: 39857599 PMCID: PMC11759860 DOI: 10.3390/biomedicines13010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Although venous thromboembolism (VTE) is the third most common cardiovascular disease, and the risk of VTE increases sharply with advancing age, approximately 40% of VTE cases are currently classified as unprovoked, highlighting the importance of risk factor research. While chronological aging is associated with the risk of VTE, the association with biological aging remains unclear. Biological aging is highly complex, influenced by several dysregulated cellular and biochemical mechanisms. In the last decade, advancements in omics methodologies provided insights into the molecular complexity of biological aging. Techniques such as high-throughput genomics, epigenomics, transcriptomics, proteomics, and metabolomics analyses identified and quantified numerous epigenetic markers, transcripts, proteins, and metabolites. These methods have also revealed the molecular alterations organisms undergo as they age. Despite the progress, there is still a lack of consensus regarding the methods for assessing and validating these biomarkers, and their application lacks standardization. This review gives an overview of biomarkers of biological aging, including telomere length, and their potential role for VTE. Furthermore, we critically examine the advantages and disadvantages of the proposed methods and discuss possible future directions for investigating biological aging in VTE.
Collapse
Affiliation(s)
| | - Cihan Ay
- Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
3
|
Pannone G, Pedicillo MC, De Stefano IS, Angelillis F, Barile R, Pannone C, Villani G, Miele F, Municinò M, Ronchi A, Serviddio G, Zito Marino F, Franco R, Colangelo T, Zamparese R. The Role of TLR-2 in Lethal COVID-19 Disease Involving Medullary and Resident Lung Megakaryocyte Up-Regulation in the Microthrombosis Mechanism. Cells 2024; 13:854. [PMID: 38786077 PMCID: PMC11120208 DOI: 10.3390/cells13100854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/27/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Patients with COVID-19 have coagulation and platelet disorders, with platelet alterations and thrombocytopenia representing negative prognostic parameters associated with severe forms of the disease and increased lethality. METHODS The aim of this study was to study the expression of platelet glycoprotein IIIa (CD61), playing a critical role in platelet aggregation, together with TRL-2 as a marker of innate immune activation. RESULTS A total of 25 patients were investigated, with the majority (24/25, 96%) having co-morbidities and dying from a fatal form of SARS-CoV-2(+) infection (COVID-19+), with 13 men and 12 females ranging in age from 45 to 80 years. When compared to a control group of SARS-CoV-2 (-) negative lungs (COVID-19-), TLR-2 expression was up-regulated in a subset of patients with deadly COVID-19 fatal lung illness. The proportion of Spike-1 (+) patients found by PCR and ISH correlates to the proportion of Spike-S1-positive cases as detected by digital pathology examination. Furthermore, CD61 expression was considerably higher in the lungs of deceased patients. In conclusion, we demonstrate that innate immune prolonged hyperactivation is related to platelet/megakaryocyte over-expression in the lung. CONCLUSIONS Microthrombosis in deadly COVID-19+ lung disease is associated with an increase in the number of CD61+ platelets and megakaryocytes in the pulmonary interstitium, as well as their functional activation; this phenomenon is associated with increased expression of innate immunity TLR2+ cells, which binds the SARS-CoV-2 E protein, and significantly with the persistence of the Spike-S1 viral sequence.
Collapse
Affiliation(s)
- Giuseppe Pannone
- Department of Clinical and Experimental Medicine, University of Foggia, Viale L.Pinto 1, 71122 Foggia, Italy; (M.C.P.); (I.S.D.S.); (F.A.)
| | - Maria Carmela Pedicillo
- Department of Clinical and Experimental Medicine, University of Foggia, Viale L.Pinto 1, 71122 Foggia, Italy; (M.C.P.); (I.S.D.S.); (F.A.)
| | - Ilenia Sara De Stefano
- Department of Clinical and Experimental Medicine, University of Foggia, Viale L.Pinto 1, 71122 Foggia, Italy; (M.C.P.); (I.S.D.S.); (F.A.)
| | - Francesco Angelillis
- Department of Clinical and Experimental Medicine, University of Foggia, Viale L.Pinto 1, 71122 Foggia, Italy; (M.C.P.); (I.S.D.S.); (F.A.)
| | - Raffaele Barile
- Department of Medical and Surgical Sciences, University of Foggia, Viale L.Pinto 1, 71122 Foggia, Italy; (R.B.); (G.S.); (T.C.)
| | - Chiara Pannone
- Faculty of Medicine, Università della Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| | - Giuliana Villani
- Policlinico Riuniti, University-Hospital, Viale L.Pinto 1, 71122 Foggia, Italy;
| | - Francesco Miele
- Department of Surgery, University of Campania “L Vanvitelli”, 80138 Naples, Italy;
| | - Maurizio Municinò
- Forensic Medicine Unit, “S. Giuliano” Hospital, Via Giambattista Basile, 80014 Giugliano in Campania, Italy;
| | - Andrea Ronchi
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L Vanvitelli”, Via Luciano Armanni, 80138 Naples, Italy; (A.R.); (F.Z.M.); (R.F.)
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, Viale L.Pinto 1, 71122 Foggia, Italy; (R.B.); (G.S.); (T.C.)
| | - Federica Zito Marino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L Vanvitelli”, Via Luciano Armanni, 80138 Naples, Italy; (A.R.); (F.Z.M.); (R.F.)
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L Vanvitelli”, Via Luciano Armanni, 80138 Naples, Italy; (A.R.); (F.Z.M.); (R.F.)
| | - Tommaso Colangelo
- Department of Medical and Surgical Sciences, University of Foggia, Viale L.Pinto 1, 71122 Foggia, Italy; (R.B.); (G.S.); (T.C.)
- Cancer Cell Signalling Unit, Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), IRCCS Fondazione Casa Sollievo della Sofferenza, Viale Cappuccini sc.c., San Giovanni Rotondo, 71013 Foggia, Italy
| | - Rosanna Zamparese
- Legal Medicine Unit, Ascoli Piceno Hospital C-G. Mazzoni, Viale Degli Iris 13, 63100 Ascoli Piceno, Italy
| |
Collapse
|
4
|
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
|
5
|
Menghoum N, Beauloye C, Lejeune S, Badii MC, Gruson D, van Dievoet MA, Pasquet A, Vancraeynest D, Gerber B, Bertrand L, Horman S, Pouleur AC. Mean platelet volume: a prognostic marker in heart failure with preserved ejection fraction. Platelets 2023; 34:2188965. [PMID: 37157842 DOI: 10.1080/09537104.2023.2188965] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is associated with high burden of comorbidities known to increase the mean platelet volume (MPV). This parameter has been associated with morbidity and mortality in HF. However, the role of platelets and the prognostic relevance of MPV in HFpEF remain largely unexplored. We aimed to evaluate the clinical usefulness of MPV as a prognostic marker in HFpEF. We prospectively enrolled 228 patients with HFpEF (79 ± 9 years; 66% females) and 38 controls of similar age and gender (78 ± 5 years; 63% females). All subjects underwent two-dimensional echocardiography and MPV measurements. Patients were followed-up for a primary end point of all-cause mortality or first HF hospitalization. The prognostic impact of MPV was determined using Cox proportional hazard models. Mean MPV was significantly higher in HFpEF patients compared with controls (MPV: 10.7 ± 1.1fL vs. 10.1 ± 1.1fL, p = .005). HFpEF patients (n = 56) with MPV >75th percentile (11.3 fL) displayed more commonly a history of ischemic cardiomyopathy. Over a median follow-up of 26 months, 136 HFpEF patients reached the composite endpoint. MPV >75th percentile was a significant predictor of the primary endpoint (HR: 1.70 [1.08; 2.67], p = .023) adjusted for NYHA class, chronic obstructive pulmonary disease, loop diuretics, renal function, and hemoglobin. We demonstrated that MPV was significantly higher in HFpEF patients compared with controls of similar age and gender. Elevated MPV was a strong and independent predictor of poor outcome in HFpEF patients and may be relevant for clinical use.
Collapse
Affiliation(s)
- Nassiba Menghoum
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Christophe Beauloye
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Sibille Lejeune
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Maria Chiara Badii
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Damien Gruson
- Clinical Biology Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | | | - Agnès Pasquet
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - David Vancraeynest
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Bernhard Gerber
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Luc Bertrand
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Anne-Catherine Pouleur
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| |
Collapse
|
6
|
Koch T, Lahu S, Coughlan JJ, Cassese S, Voll F, Ndrepepa G, Menichelli M, Valina C, Hemetsberger R, Witzenbichler B, Bernlochner I, Joner M, Xhepa E, Mayer K, Kessler T, Laugwitz KL, Richardt G, Schunkert H, Angiolillo DJ, Sibbing D, Kastrati A, Kufner S. Association between Platelet Count and Treatment Effect of Ticagrelor or Prasugrel in Patients with Acute Coronary Syndromes. Thromb Haemost 2023; 123:464-477. [PMID: 36442805 PMCID: PMC10060058 DOI: 10.1055/a-1988-5047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 10/27/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND The relative efficacy and safety of ticagrelor and prasugrel based dual antiplatelet therapy strategies according to the platelet count (PC) in patients with acute coronary syndromes (ACS) have not been defined. METHODS This is a posthoc analysis of the ISAR-REACT 5 trial, in which patients presenting with ACS were randomized to treatment with ticagrelor versus prasugrel. Patients were divided into quartiles according to PC. The primary endpoint was incidence of death, myocardial infarction, or stroke, and the safety endpoint was incidence of BARC (Bleeding Academic Research Consortium) type 3 to 5 bleeding at 12 months. RESULTS A total of 3,943 patients with known PC (997 patients in quartile 1 (Q1), 1,003 in quartile 2 (Q2) [205 ± 10.3 × 109/L], 961 patients in quartile 3 (Q3) [241 ± 11.7 × 109/L], and 982 patients in quartile 4 (Q4) [317 ± 68.6 × 109/L]). There was no significant interaction between treatment arm (ticagrelor vs. prasugrel) and PC group with respect to primary endpoint (Q1: 8.8 vs. 6.3%, hazard ratio [HR] =1.41, 95% confidence interval [CI]: 0.89-2.23; p = 0.148; Q2: 9.9 vs. 5.8%, HR = 1.68, 95% CI: 1.06-2.66; p = 0.027; Q3: 7.8 vs. 5.5%, HR = 1.43, 95% CI: 0.87-2.37; p = 0.159; Q4: 10.1 vs. 10.1%, HR = 1.05, 95% CI: 0.71-1.57; p = 0.799; p for interaction [p int] = 0.482) and with respect to bleeding endpoint (Q1: 5.8 vs. 4.2%, HR = 1.41, 95% CI: 0.76-2.63; p = 0.279; Q2: 6.4 vs. 3.7%, HR = 1.62, 95% CI: 0.85-2.06; p = 0.140; Q3: 4.4 vs. 3.0%, HR = 1.53, 95% CI: 0.73-3.18; p = 0.258; Q4: 5.6 vs. 8.5%, HR = 0.67, 95% CI: 0.40-1.14; p = 0.138, p int = 0.102). CONCLUSIONS In this analysis, incidences of ischemic and bleeding events at 12 months are comparable across quartiles of platelet count.
Collapse
Affiliation(s)
- Tobias Koch
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Shqipdona Lahu
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - J. J. Coughlan
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Salvatore Cassese
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Felix Voll
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Gjin Ndrepepa
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | | | - Christian Valina
- Department of Cardiology and Angiology II, University Heart Center Freiburg - Bad Krozingen, Standort Bad Krozingen, Germany
| | - Rayyan Hemetsberger
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | | | - Isabell Bernlochner
- Medizinische Klinik und Poliklinik Innere Medizin I (Kardiologie, Angiologie, Pneumologie), Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Michael Joner
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Erion Xhepa
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Katharina Mayer
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Thorsten Kessler
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Karl-Ludwig Laugwitz
- Medizinische Klinik und Poliklinik Innere Medizin I (Kardiologie, Angiologie, Pneumologie), Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Gert Richardt
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Heribert Schunkert
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Dominick J. Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Dirk Sibbing
- Privatklinik Lauterbacher Mühle am Ostersee, Iffeldorf and Ludwig-Maximilians-Universität München, Munich, Germany
| | - Adnan Kastrati
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Sebastian Kufner
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
7
|
Sethi A, Melamud E. Joint inference of physiological network and survival analysis identifies factors associated with aging rate. CELL REPORTS METHODS 2022; 2:100356. [PMID: 36590696 PMCID: PMC9795372 DOI: 10.1016/j.crmeth.2022.100356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/11/2022] [Accepted: 11/10/2022] [Indexed: 12/04/2022]
Abstract
We describe methodology for joint reconstruction of physiological-survival networks from observational data capable of identifying key survival-associated variables, inferring a minimal physiological network structure, and bridging this network to the Gompertzian survival layer. Using synthetic network structures, we show that the method is capable of identifying aging variables in cohorts as small as 5,000 participants. Applying the methodology to the observational human cohort, we find that interleukin-6, vascular calcification, and red-blood distribution width are strong predictors of baseline fitness. More important, we find that red blood cell counts, kidney function, and phosphate level are directly linked to the Gompertzian aging rate. Our model therefore enables discovery of processes directly linked to the aging rate of our species. We further show that this epidemiological framework can be applied as a causal inference engine to simulate the effects of interventions on physiology and longevity.
Collapse
Affiliation(s)
- Anurag Sethi
- Calico Life Sciences LLC, 1170 Veterans Blvd., South San Francisco, CA 94080, USA
| | - Eugene Melamud
- Calico Life Sciences LLC, 1170 Veterans Blvd., South San Francisco, CA 94080, USA
| |
Collapse
|
8
|
Caulis Polygoni Multiflori Accelerates Megakaryopoiesis and Thrombopoiesis via Activating PI3K/Akt and MEK/ERK Signaling Pathways. Pharmaceuticals (Basel) 2022; 15:ph15101204. [PMID: 36297316 PMCID: PMC9607024 DOI: 10.3390/ph15101204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/19/2022] [Accepted: 09/24/2022] [Indexed: 11/23/2022] Open
Abstract
Thrombocytopenia is one of the most common complications of cancer therapy. Until now, there are still no satisfactory medications to treat chemotherapy and radiation-induced thrombocytopenia (CIT and RIT, respectively). Caulis Polygoni Multiflori (CPM), one of the most commonly used Chinese herbs, has been well documented to nourish blood for tranquilizing the mind and treating anemia, suggesting its beneficial effect on hematopoiesis. However, it is unknown whether CPM can accelerate megakaryopoiesis and thrombopoiesis. Here, we employ a UHPLC Q–Exactive HF-X mass spectrometer (UHPLC QE HF-X MS) to identify 11 ingredients in CPM. Then, in vitro experiments showed that CPM significantly increased megakaryocyte (MK) differentiation and maturation but did not affect apoptosis and lactate dehydrogenase (LDH) release of K562 and Meg-01 cells. More importantly, animal experiments verified that CPM treatment markedly accelerated platelet recovery, megakaryopoiesis and thrombopoiesis in RIT mice without hepatic and renal toxicities in vivo. Finally, RNA-sequencing (RNA-seq) and western blot were used to determine that CPM increased the expression of proteins related to PI3K/Akt and MEK/ERK (MAPK) signaling pathways. On the contrary, blocking PI3K/Akt and MEK/ERK signaling pathways with their specific inhibitors suppressed MK differentiation induced by CPM. In conclusion, for the first time, our study demonstrates that CPM may be a promised thrombopoietic agent and provide an experimental basis for expanding clinical use.
Collapse
|
9
|
Zhao L, Liu M, Liu L, Guo W, Yang H, Chen S, Yu J, Li M, Fang Q, Lai X, Yang L, Zhang X. The association of co-exposure to polycyclic aromatic hydrocarbon and phthalates with blood cell-based inflammatory biomarkers in children: A panel study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 307:119479. [PMID: 35598818 DOI: 10.1016/j.envpol.2022.119479] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 06/15/2023]
Abstract
The association of co-exposure to polycyclic aromatic hydrocarbons (PAHs) and phthalates (PAEs) with blood cell-based inflammatory biomarkers is largely unknown. We conducted a panel study of 144 children aged 4-12 years, with up to 3 repeated visits across 3 seasons. For each visit, we collected the first-morning urine for 4 consecutive days and fasting blood on the day of physical examination. We developed a gas chromatography/tandem mass spectrometry method to detect the metabolites of 10 PAHs (OH-PAHs) and 10 PAEs (mPAEs) in urine samples. We employed linear mixed-effects models to evaluate the individual associations of each OH-PAH and mPAE with blood cell-based inflammatory biomarkers over different lag times. Bayesian kernel machine regression (BKMR) and quantile g-computation were used to evaluate the overall associations of OH-PAHs and mPAEs mixtures with blood cell-based inflammatory biomarkers. After multiple adjustments, we found positive associations of summed hydroxylphenanthrene (∑OHPHE), summed OH-PAHs, and mono-n-butyl phthalate with inflammatory biomarkers such as neutrophil count, neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and the systemic immune-inflammation index (SII) at lag 0 (the day of physical examination). Each 1% increase in ∑OHPHE was related to a 0.18% (95% confidence interval: 0.10%, 0.25%) increase in SII, which was the strongest among the above associations. The results of BKMR and quantile g-computation suggested that co-exposure to PAHs and PAEs mixture was associated with an elevated white blood cell count, neutrophil count, neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and SII, to which ∑OHPHE and 1-hydroxypyrene (1-OHPYR) might be the major contributors. In addition, gender and age modified the associations of ∑OHPHE and 1-OHPYR with inflammatory biomarkers, where girls and younger children were more susceptible. In conclusion, co-exposure to PAHs and PAEs was associated with elevated inflammation in children, in which ∑OHPHE and 1-OHPYR might play important roles.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Public Health, Medical College of Qinghai University, Xining, Qinghai, China
| | - Miao Liu
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Linlin Liu
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenting Guo
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huihua Yang
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuang Chen
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Yu
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meng Li
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Fang
- Department of Medical Affairs, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, China
| | - Xuefeng Lai
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liangle Yang
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
10
|
von Stemann JH, Pedersen OBV, Hjalgrim H, Erikstrup C, Ullum H, Dowsett J, Thørner LW, Larsen MAH, Sørensen E, Hansen MB, Ostrowski SR. IL-6 Autoantibodies Predict Lower Platelet Counts and Altered Plasma Cytokine Profiles in Healthy Blood Donors: Results From the Danish Blood Donor Study. Front Med (Lausanne) 2022; 9:914262. [PMID: 35814772 PMCID: PMC9263719 DOI: 10.3389/fmed.2022.914262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/02/2022] [Indexed: 12/03/2022] Open
Abstract
Cytokine-specific autoantibodies (c-aAb) represent a novel type of immune dysfunction. Though they have been detected in both patient cohorts and healthy individuals, and have immunomodulatory properties, the full extent of their influence remains unknown. Based on the critical role of several cytokines in thrombopoiesis, we investigated if there is an association between c-aAb and platelet variables in healthy individuals, with a specific focus on c-aAb against a known thrombopoietic cytokine, IL-6. Using platelet count and mean platelet volume in 3,569 healthy participants of the Danish Blood Donor Study as dependent variables, we performed a series of multivariate regression analyses using five cytokine autoantibodies, including IL-6 c-aAb, as independent variables. In men, high titers of IL-6 c-aAb were negatively associated with platelet counts (β = −24 *109/l (95% confidence interval −43 to −6), p = 0.008) and positively associated with mean platelet volume (β = 0.4 fL (95% confidence interval 0.0–0.7) p = 0.043). These associations were exacerbated when adjusting for undetectable C-reactive protein levels, which we used as a proxy for c-aAb mediated IL-6 inhibition in vivo. Furthermore, in a smaller subgroup, individuals with high vs. low titer IL-6 c-aAb had different profiles of plasma IL-6, IL-10, TNFα and TPO, further suggesting a functional inhibition of IL-6 by high titers of circulating IL-6 c-aAb. We therefore speculate that in addition to their immunomodulatory potential IL-6 c-aAb may interfere with thrombopoiesis – directly or indirectly – under normal physiological conditions. This study is the first to suggest an influence of c-aAb on platelets in healthy individuals, beyond their apparent effects on immune competence.
Collapse
Affiliation(s)
- Jakob Hjorth von Stemann
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- *Correspondence: Jakob Hjorth von Stemann
| | - Ole Birger Vesterager Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Hjalgrim
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Centre for Cancer Research, Danish Cancer Society, Copenhagen, Denmark
- Department of Haematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Joseph Dowsett
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lise Wegner Thørner
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Margit Anita Hørup Larsen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Morten Bagge Hansen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Huang Y, Hong M, Qu Z, Zheng W, Hu H, Li L, Lu T, Xie Y, Ying S, Zhu Y, Liu L, Huang W, Fu S, Chen J, Wu K, Liu M, Luo Q, Wu Y, He F, Zhang J, Zhang J, Chen Y, Zhao M, Cai Z, Huang H, Sun J. Non-Ablative Chemotherapy Followed by HLA-Mismatched Allogeneic CD3 + T-Cells Infusion Causes An Augment of T-Cells With Mild CRS: A Multi-Centers Single-Arm Prospective Study on Elderly Acute Myeloid Leukemia and int-2/High Risk Myelodysplastic Syndrome Patients. Front Oncol 2021; 11:741341. [PMID: 34722293 PMCID: PMC8548743 DOI: 10.3389/fonc.2021.741341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Objective To evaluate the efficacy and safety of standard or low-dose chemotherapy followed by HLA-mismatched allogeneic T-cell infusion (allo-TLI) for the treatment of elderly patients with acute myeloid leukemia (AML) and patients with intermediate-2 to high-risk myelodysplastic syndrome (MDS). Methods We carried out a prospective, multicenter, single-arm clinical trial. Totally of 25 patients were enrolled, including 17 AML patients and 8 MDS patients. Each patient received four courses of non-ablative chemotherapy, with HLA-mismatched donor CD3+ allo-TLI 24 h after each course. AML patients received chemotherapy with decitabine, idarubicin, and cytarabine, and MDS patients received decitabine, cytarabine, aclarubicin, and granulocyte colony-stimulating factor. Results A total of 79 procedures were performed. The overall response rates of the AML and MDS patients were 94% and 75% and the 1-year overall survival rates were 88% (61-97%) and 60% (13-88%), respectively. The overall 60-day treatment-related mortality was 8%. Compared with a historical control cohort that received idarubicin plus cytarabine (3 + 7), the study group showed significantly better overall response (94% vs. 50%, P=0.002) and overall survival rates (the 1-year OS rate was 88% vs. 27%, P=0.014). Post-TLI cytokine-release syndrome (CRS) occurred after 79% of allo-TLI operations, and 96% of CRS reactions were grade 1. Conclusion Elderly AML patients and intermediate-2 to high-risk MDS patients are usually insensitive to or cannot tolerate regular chemotherapies, and may not have the opportunity to undergo allogeneic stem cell transplantation. Our study showed that non-ablative chemotherapy followed by HLA-mismatched allo-TLI is safe and effective, and may thus be used as a first-line treatment for these patients. Clinical Trial Registration https://www.chictr.org.cn/showproj.aspx?proj=20112.
Collapse
Affiliation(s)
- Yan Huang
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minghua Hong
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhigang Qu
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Weiyan Zheng
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huixian Hu
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Linjie Li
- Department of Hematology, The Central Hospital of Lishui City, Lishui, China
| | - Ting Lu
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Xie
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuangwei Ying
- Department of Hematology, Taizhou Hospital of Zhejiang Province, Taizhou, China
| | - Yuanyuan Zhu
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lizhen Liu
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weijia Huang
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shan Fu
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jin Chen
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Kangli Wu
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Mingsuo Liu
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Qiulian Luo
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Yajun Wu
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Fang He
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jingcheng Zhang
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Junyu Zhang
- Department of Hematology, The Central Hospital of Lishui City, Lishui, China
| | - Yu Chen
- Department of Hematology, The Central Hospital of Lishui City, Lishui, China
| | - Minlei Zhao
- Department of Hematology, The Central Hospital of Lishui City, Lishui, China
| | - Zhen Cai
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - He Huang
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Sun
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Chen X, Wang C, Sun N, Pan S, Li R, Li X, Zhao J, Tong H, Tang Y, Han J, Qiao J, Qiu H, Wang H, Yang J, Ikezoe T. Aurka loss in CD19 + B cells promotes megakaryocytopoiesis via IL-6/STAT3 signaling-mediated thrombopoietin production. Theranostics 2021; 11:4655-4671. [PMID: 33754019 PMCID: PMC7978311 DOI: 10.7150/thno.49007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 02/15/2021] [Indexed: 01/21/2023] Open
Abstract
Rationale: Aurora kinase A (Aurora-A), which is required for mitosis, is a therapeutic target in various tumors. Targeting Aurora-A led to an increase in the differentiation and polyploidization of megakaryocytes both in vivo and in vitro. However, the mechanisms involved in controlling megakaryocyte differentiation have not been fully elucidated. Methods: Conditional Aurka knockout mice were generated. B cell development, platelet development and function were subsequently examined. Proplatelet formation, in vivo response to mTPO, post-transfusion experiment, colony assay, immunofluorescence staining and quantification, and ChIP assay were conducted to gain insights into the mechanisms of Aurka loss in megakaryocytopoiesis. Results: Loss of Aurka in CD19+ B cells impaired B cell development in association with an increase in the number of platelets in peripheral blood (PB). Surprisingly, thrombopoietin (TPO) production and IL-6 were elevated in the plasma in parallel with an increase in the number of differentiated megakaryocytes in the bone marrow (BM) of Aurkaf/f;Cd19Cre/+ mice. Interestingly, compared with that of the Aurkaf/f mice, a higher number of CD19+ B cells close to megakaryocytes was observed in the BM of the Aurkaf/f;Cd19Cre/+ mice. Moreover, Aurka loss in CD19+ B cells induced signal transducer and activator of transcription-3 (STAT3) activation. Inhibition of STAT3 reduced the Tpo mRNA levels. ChIP assays revealed that STAT3 bound to the TPO promoter. Additionally, STAT3-mediated TPO transcription was an autocrine effect provoked by IL-6, at least partially. Conclusions: Deletion of Aurka in CD19+ B cells led to an increase in IL-6 production, promoting STAT3 activation, which in turn contributed to TPO transcription and megakaryocytopoiesis.
Collapse
|
13
|
Ding S, Wang M, Fang S, Xu H, Fan H, Tian Y, Zhai Y, Lu S, Qi X, Wei F, Sun G, Sun X. D-dencichine Regulates Thrombopoiesis by Promoting Megakaryocyte Adhesion, Migration and Proplatelet Formation. Front Pharmacol 2018; 9:297. [PMID: 29666579 PMCID: PMC5891617 DOI: 10.3389/fphar.2018.00297] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 03/15/2018] [Indexed: 01/09/2023] Open
Abstract
Life-threatening chemotherapy-induced thrombocytopenia can increase the risk of bleeding due to a dramatic low platelet count, which may limit or delay treatment schedules in cancer patients. The pressing need for the rapid alleviation of the symptoms of thrombocytopenia has prompted us to search for novel highly effective and safe thrombopoietic agents. Pharmacological investigations have indicated that dencichine can prevent and treat blood loss and increase the number of platelets. On the basis of the neurotoxicity of dencichine, D-dencichine is artificially synthesized in the laboratory. Our initial results showed that D-dencichine had potential to elevate peripheral platelet levels in mice with carboplatin-induced thrombocytopenia. However, the mechanisms of D-dencichine on thrombopoiesis have been poorly understood. In this study, we found that sequential administration of D-dencichine had a distinct ability to elevate numbers of reticulated platelets, and did not alter their clearance. Moreover, we demonstrated that D-dencichine was able to modulate the return of hematopoietic factors to normal levels, including thrombopoietin and IL-6. However, subsequent analysis revealed that D-dencichine treatment had no direct effects on megakaryocytes proliferation, differentiation, and polyploidization. Further in vitro studies, we demonstrated for the first time that D-dencichine significantly stimulated megakaryocyte adhesion, migration, and proplatelet formation in a dose-dependent manner through extracellular regulated protein kinases1/2 (ERK1/2) and v-akt murine thymoma viral oncogene homolog (AKT) signaling pathways. This study sufficiently characterized the role of the effects of D-dencichine treatment on the regulation of thrombopoiesis and provided a promising avenue for CIT treating.
Collapse
Affiliation(s)
- Shilan Ding
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Song Fang
- Kunming Shenghuo Pharmaceutical Group Co., Ltd., Kunming, China
| | - Huibo Xu
- Academy of Chinese Medical Sciences of Jilin Province, Jilin, China
| | - Huiting Fan
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Tian
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Yadong Zhai
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Shan Lu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xin Qi
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fei Wei
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Čolak E, Ignjatović S, Radosavljević A, Žorić L. The association of enzymatic and non-enzymatic antioxidant defense parameters with inflammatory markers in patients with exudative form of age-related macular degeneration. J Clin Biochem Nutr 2017; 60:100-107. [PMID: 28366988 PMCID: PMC5371514 DOI: 10.3164/jcbn.16-30] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 06/26/2016] [Indexed: 02/05/2023] Open
Abstract
There are evidence that oxidative stress and inflammation are involved in the pathogenesis of the age-related macular degeneration (AMD). The aim of this study was to analyze the antioxidant defense parameters and inflammatory markers in patients with exudative form of AMD (eAMD), their mutual correlations and association with the specific forms of AMD. The cross-sectional study, included 75 patients with the eAMD, 31 patients with the early form, and 87 aged-matched control subjects. Significantly lower SOD, TAS and albumin values and higher GR, CRP and IL-6 were found in the eAMD compared to the early form (p<0.05). Significant negative correlations were found between GPx and fibrinogen (r = -0.254), TAS and IL-6 (r = -0.999) and positive correlations between uric acid and CRP (r = 0.292), IL-6 and uric acid (r = 0.398) in the eAMD. A significant association of CRP (OR: 1.16, 95% CI: 1.03-1.32, p = 0.018), fibrinogen (OR: 2.21, 95% CI: 1.14-4.85, p = 0.021), TAS (OR: 7.45, 95% CI: 3.97-14.35, p = 0.0001), albumin (OR: 1.25, 95% CI: 1.11-1.41, p = 0.0001) and uric acid (OR: 1.006, 95% CI: 1.00-1.02, p = 0.003) was found with the eAMD. In conclusion it may be suggested, there is a significant impairment of antioxidant and inflammatory parameter levels in eAMD patients. In addition, significant association exists between the tested inflammatory markers and antioxidant parameters with late-eAMD.
Collapse
Affiliation(s)
- Emina Čolak
- Institute of Medical Biochemistry, Clinical Center of Serbia, School of Pharmacy, University of Belgrade, Belgrade 11000, Serbia
| | - Svetlana Ignjatović
- Institute of Medical Biochemistry, Clinical Center of Serbia, School of Pharmacy, University of Belgrade, Belgrade 11000, Serbia
| | - Aleksandra Radosavljević
- Institute of Ophthalmology, Medical Retina Department, Clinical Center of Serbia, School of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Lepša Žorić
- Clinic for Eye Diseases, Clinical Center, Faculty of Medicine, Settlement Kosovska Mitrovica, Kosovska Mitrovica 38200, Serbia
| |
Collapse
|
15
|
IL-6 and MCP-1 genetic polymorphisms are predictive of decreased platelet counts caused by chemoradiotherapy in esophageal cancer. Esophagus 2016. [DOI: 10.1007/s10388-016-0522-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
16
|
Senchenkova EY, Komoto S, Russell J, Almeida-Paula LD, Yan LS, Zhang S, Granger DN. Interleukin-6 mediates the platelet abnormalities and thrombogenesis associated with experimental colitis. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:173-81. [PMID: 23673000 DOI: 10.1016/j.ajpath.2013.03.014] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Revised: 02/25/2013] [Accepted: 03/20/2013] [Indexed: 01/09/2023]
Abstract
Clinical studies and animal experimentation have shown that colonic inflammation is associated with an increased number and reactivity of platelets, coagulation abnormalities, and enhanced thrombus formation. The objective of this study was to define the contribution of IL-6 to the thrombocytosis, exaggerated agonist-induced platelet aggregation, and enhanced extra-intestinal thrombosis that occur during experimental colitis. The number of mature and immature platelets, platelet life span, thrombin-induced platelet aggregation response, and light/dye-induced thrombus formation in cremaster muscle arterioles were measured in wild-type (WT) and IL-6-deficient (IL-6(-/-)) mice with dextran sodium sulfate (DSS)-induced colitis. DSS colitis in WT mice was associated with thrombocytosis with an elevated number of both mature and immature platelets and no change in platelet life span. The thrombocytosis response was absent in IL-6(-/-) mice. DSS treatment also enhanced the platelet aggregation response to thrombin and accelerated thrombus development in WT mice, but not in IL-6(-/-) mice. Exogenous IL-6 administered to WT mice elicited a dose-dependent enhancement of thrombus formation. These findings indicate that IL-6 mediates the thrombocytosis, platelet hyperreactivity, and accelerated thrombus development associated with experimental colitis. The IL-6-dependent colitis-induced thrombocytosis appears to result from an enhancement of thrombopoiesis because platelet life span is unchanged.
Collapse
Affiliation(s)
- Elena Y Senchenkova
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Iwaki T, Urano T, Umemura K. PAI-1, progress in understanding the clinical problem and its aetiology. Br J Haematol 2012; 157:291-8. [PMID: 22360729 DOI: 10.1111/j.1365-2141.2012.09074.x] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/24/2012] [Indexed: 01/07/2023]
Abstract
Plasminogen activator inhibitor-1 (PAI-1, also known as SERPINE1) is a member of the serine protease inhibitor (SERPIN) superfamily and is the primary physiological regulator of urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA) activity. Although the principal function of PAI-1 is the inhibition of fibrinolysis, PAI-1 possesses pleiotropic functions besides haemostasis. In the quarter century since its discovery, a number of studies have focused on improving our understanding of PAI-1 functions in vivo and in vitro. The use of Serpine1-deficient mice has particularly enhanced our understanding of the functions of PAI-1 in various physiological and pathophysiological conditions. In this review, the results of recent studies on PAI-1 and its role in clinical conditions are discussed.
Collapse
Affiliation(s)
- Takayuki Iwaki
- Department of Pharmacology, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | | | | |
Collapse
|
18
|
Cesari M, Pahor M, Incalzi RA. Plasminogen activator inhibitor-1 (PAI-1): a key factor linking fibrinolysis and age-related subclinical and clinical conditions. Cardiovasc Ther 2010; 28:e72-91. [PMID: 20626406 PMCID: PMC2958211 DOI: 10.1111/j.1755-5922.2010.00171.x] [Citation(s) in RCA: 318] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION The close relationship existing between aging and thrombosis has growingly been studied in this last decade. The age-related development of a prothrombotic imbalance in the fibrinolysis homeostasis has been hypothesized as the basis of this increased cardiovascular and cerebrovascular risk. Fibrinolysis is the result of the interactions among multiple plasminogen activators and inhibitors constituting the enzymatic cascade, and ultimately leading to the degradation of fibrin. The plasminogen activator system plays a key role in a wide range of physiological and pathological processes. METHODS Narrative review. RESULTS Plasminogen activator inhibitor-1 (PAI-1) is a member of the superfamily of serine-protease inhibitors (or serpins), and the principal inhibitor of both the tissue-type and the urokinase-type plasminogen activator, the two plasminogen activators able to activate plasminogen. Current evidence describing the central role played by PAI-1 in a number of age-related subclinical (i.e., inflammation, atherosclerosis, insulin resistance) and clinical (i.e., obesity, comorbidities, Werner syndrome) conditions is presented. CONCLUSIONS Despite some controversial and unclear issues, PAI-1 represents an extremely promising marker that may become a biological parameter to be progressively considered in the prognostic evaluation, in the disease monitoring, and as treatment target of age-related conditions in the future.
Collapse
Affiliation(s)
- Matteo Cesari
- Area di Geriatria, Università Campus Bio-Medico, Rome, Italy.
| | | | | |
Collapse
|
19
|
Eidelman O, Jozwik C, Huang W, Srivastava M, Rothwell SW, Jacobowitz DM, Ji X, Zhang X, Guggino W, Wright J, Kiefer J, Olsen C, Adimi N, Mueller GP, Pollard HB. Gender dependence for a subset of the low-abundance signaling proteome in human platelets. HUMAN GENOMICS AND PROTEOMICS : HGP 2010; 2010:164906. [PMID: 20981232 PMCID: PMC2958630 DOI: 10.4061/2010/164906] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 01/05/2010] [Indexed: 11/23/2022]
Abstract
The incidence of cardiovascular diseases is ten-times higher in males than females, although the biological basis for this gender disparity is not known. However, based on the fact that antiplatelet drugs are the mainstay for prevention and therapy, we hypothesized that the signaling proteomes in platelets from normal male donors might be more activated than platelets from normal female donors. We report here that platelets from male donors express significantly higher levels of signaling cascade proteins than platelets from female donors. In silico connectivity analysis shows that the 24 major hubs in platelets from male donors focus on pathways associated with megakaryocytic expansion and platelet activation. By contrast, the 11 major hubs in platelets from female donors were found to be either negative or neutral for platelet-relevant processes. The difference may suggest a biological mechanism for gender discrimination in cardiovascular disease.
Collapse
Affiliation(s)
- Ofer Eidelman
- Department of Anatomy, Physiology and Genetics, USU Center for Medical Proteomics, Uniformed Services University, School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Catherine Jozwik
- Department of Anatomy, Physiology and Genetics, USU Center for Medical Proteomics, Uniformed Services University, School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Wei Huang
- Department of Anatomy, Physiology and Genetics, USU Center for Medical Proteomics, Uniformed Services University, School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Meera Srivastava
- Department of Anatomy, Physiology and Genetics, USU Center for Medical Proteomics, Uniformed Services University, School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Stephen W. Rothwell
- Department of Anatomy, Physiology and Genetics, USU Center for Medical Proteomics, Uniformed Services University, School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - David M. Jacobowitz
- National Institute for Mental Health, NIH, 9500 Rockville Pike, Bethesda, MD 20892, USA
| | - Xiaoduo Ji
- Department of Anatomy, Physiology and Genetics, USU Center for Medical Proteomics, Uniformed Services University, School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Xiuying Zhang
- Department of Anatomy, Physiology and Genetics, USU Center for Medical Proteomics, Uniformed Services University, School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - William Guggino
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jerry Wright
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jeffrey Kiefer
- Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Cara Olsen
- Department of Preventive Medicine and Biometrics, Uniformed Services University School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Nima Adimi
- Department of Anatomy, Physiology and Genetics, USU Center for Medical Proteomics, Uniformed Services University, School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Gregory P. Mueller
- Department of Anatomy, Physiology and Genetics, USU Center for Medical Proteomics, Uniformed Services University, School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Harvey B. Pollard
- Department of Anatomy, Physiology and Genetics, USU Center for Medical Proteomics, Uniformed Services University, School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
20
|
Breimo ES, Østerud B. Studies of biological functions in blood cells from individuals with large platelets. Platelets 2009; 14:413-9. [PMID: 14713510 DOI: 10.1080/02697450310001632597] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The present study was performed to explore differences in the expression of P-selectin and IL-8 production of blood cells in healthy individuals with large size platelets (MI-families) as compared to people having normal size platelets. A positive correlation between LPS-induced IL-8 production per platelet in whole blood and mean platelet volume (MPV) was found in the large platelet group (R=0.74, P<0.05). When the large and normal groups were combined the correlation was nearly, but not quite significant (R=0.46, P<0.06). There was also a positive correlation between sP-selectin and MPV (R=0.42, P<0.05). Furthermore, IL-8 in serum was positively correlated to sP-selectin in serum (R=0.68, P<0.005). sP-selectin baseline values in citrated plasma correlated significantly with values found in serum (R=0.72, P<0.0005), indicating that sP-selectin in blood originates from the platelets rather than from endothelial cells. Significant correlations were also found in both groups between P-selectin and CD40L (R=0.44, P<0.05) and P-selectin and RANTES (R=0.44, P<0.05). A significant correlation was also found between PDGF and RANTES (R=0.44, P<0.05). Our results suggest that larger platelets enhance the production of IL-8 more than normally sized platelets. This phenomenon is probably mediated through P-selectin exposed on platelets.
Collapse
Affiliation(s)
- Einar S Breimo
- Department of Biochemistry, Institute of Medical Biology, University of Tromsø, Norway.
| | | |
Collapse
|
21
|
Low-dose acetylsalicylic acid inhibits the secretion of interleukin-6 from white adipose tissue. Int J Obes (Lond) 2008; 32:1807-15. [PMID: 18982014 DOI: 10.1038/ijo.2008.190] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Chronically elevated interleukin-6 (IL-6) is implicated in obesity-associated pathologies, where a proportion of this cytokine is derived from adipose tissue. Proinflammatory prostaglandins, which regulate this cytokine elsewhere, are also produced by this tissue. OBJECTIVE To investigate whether constitutively active cyclooxygenase (COX)/prostaglandin (PG) pathway in white adipose tissue (WAT) is responsible for basal IL-6 production. DESIGN The effect of acetylsalicylic acid (ASA), an inhibitor of COX, on IL-6 was assessed in human subjects and mice. COX, downstream PG synthase (PGS) activity and PG receptor signalling were determined in subcutaneous (SC), gonadal (GN) WAT and adipocytes. METHODS AND RESULTS In obese humans, low-dose ASA (150 mg day(-1) for 10 days) inhibited systemic IL-6 and reduced IL-6 release from SC WAT ex vivo (0.2 mM). Similarly, in mice, ASA (0.2 and 2.0 mg kg(-1)) suppressed SC WAT 6-keto-PGF(1alpha) (a stable metabolite of prostacyclin) and IL-6 release. Although both COX isoforms are comparably expressed, prostacyclin synthase expression is higher in GN WAT, with levels of activity correlating directly with IL-6. Both ASA (5 mM) and NS-398 (COX-2 selective inhibitor <or=1 microM), but not SC-560 (COX-1 selective inhibitor <or=1 microM), attenuated IL-6 release from murine WAT in vitro and abolished its depot differences. Prostacyclin receptor (IP) and, to a lesser extent, PGE(2) (EP2 and EP4) receptor agonists elevated the release of IL-6 from adipocytes. CONCLUSIONS In adipose tissue, constitutive COX-2-coupled prostacyclin triggers the release of basal IL-6, which in obese subjects is significantly dampened by ASA ingestion, thus offering a novel, modifiable pathway to regulate the potentially pathological component of this cytokine.
Collapse
|
22
|
Omoigui S. The Interleukin-6 inflammation pathway from cholesterol to aging--role of statins, bisphosphonates and plant polyphenols in aging and age-related diseases. Immun Ageing 2007; 4:1. [PMID: 17374166 PMCID: PMC1845171 DOI: 10.1186/1742-4933-4-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 03/20/2007] [Indexed: 12/25/2022]
Abstract
We describe the inflammation pathway from Cholesterol to Aging. Interleukin 6 mediated inflammation is implicated in age-related disorders including Atherosclerosis, Peripheral Vascular Disease, Coronary Artery Disease, Osteoporosis, Type 2 Diabetes, Dementia and Alzheimer's disease and some forms of Arthritis and Cancer. Statins and Bisphosphonates inhibit Interleukin 6 mediated inflammation indirectly through regulation of endogenous cholesterol synthesis and isoprenoid depletion. Polyphenolic compounds found in plants, fruits and vegetables inhibit Interleukin 6 mediated inflammation by direct inhibition of the signal transduction pathway. Therapeutic targets for the control of all the above diseases should include inhibition of Interleukin-6 mediated inflammation.
Collapse
Affiliation(s)
- Sota Omoigui
- Division of Inflammation and Pain Medicine, LA Pain Clinic, 4019 W Rosecrans Ave, Los Angeles, CA 90250, USA.
| |
Collapse
|
23
|
Abstract
Coagulation is triggered by inflammatory mediators in a number of ways. However, to prevent unwanted clot formation, several natural anticoagulant mechanisms exist, such as the antithrombin-heparin mechanism, the tissue factor pathway inhibitor mechanism and the protein C anticoagulant pathway. This review examines the ways in which these pathways are down-regulated by inflammation, thus limiting clot formation and decreasing the natural anti-inflammatory mechanisms that these pathways possess.
Collapse
Affiliation(s)
- C T Esmon
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Howard Hughes Medical Institute, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
24
|
Heseltine JC, Panciera DL, Troy GC, Monroe WE, Brooks MB, Feldman BF. Effect of Levothyroxine Administration on Hemostatic Analytes in Doberman Pinschers with von Willebrand Disease. J Vet Intern Med 2005. [DOI: 10.1111/j.1939-1676.2005.tb02721.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
25
|
Abstract
Diabetes is a well-recognised risk factor for atherosclerotic cardiovascular disease and in fact most diabetic patients die from vascular complications. The Diabetes Control and Complications Trial (DCCT) and the U.K. Prospective Diabetes Study (UKPDS) indicate a consistent relationship between hyperglycaemia and the incidence of chronic vascular complications in patients with diabetes. Platelets are essential for haemostasis, and abnormalities of platelet function may cause vascular disease in diabetes. Diabetic patients have hyperreactive platelets with exaggerated adhesion, aggregation and thrombin generation. In summary, the entire coagulation cascade is dysfunctional in diabetes. This review provides a comprehensive overview of the physiological role of platelets in maintaining haemostasis and of the pathophysiological processes that contribute to platelet dysfunction in diabetes and associated cardiovascular diseases, with special emphasis on proteomic approaches and leukocyte-platelet cross-talk.
Collapse
Affiliation(s)
- Bernd Stratmann
- Herz- und Diabeteszentrum NRW, Georgstral3e 11, 32545 Bad Oeynhausen, Germany
| | | |
Collapse
|
26
|
Bernardo A, Ball C, Nolasco L, Moake JF, Dong JF. Effects of inflammatory cytokines on the release and cleavage of the endothelial cell-derived ultralarge von Willebrand factor multimers under flow. Blood 2004; 104:100-6. [PMID: 15026315 DOI: 10.1182/blood-2004-01-0107] [Citation(s) in RCA: 408] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
ADAMTS13 cleaves ultralarge and hyperreactive von Willebrand factor (ULVWF) freshly released from activated endothelial cells to smaller and less active forms. This process may be affected by the amount of ULVWF released and the processing capacity of ADAMTS13, contributing to the development of thrombotic diseases. We examined the effects of inflammatory cytokines on the release and cleavage of ULVWF to evaluate potential links between inflammation and thrombosis. Human umbilical vein endothelial cells were treated with interleukin 6 (IL-6), IL-8, or tumor necrosis factor alpha (TNF-alpha), and the formation of platelet-decorated ULVWF strings was quantitated. IL-8 and TNF-alpha significantly stimulated the release of ULVWF in a dose-dependent manner. IL-6 induced ULVWF release only when it was in complex with the soluble IL-6 receptor. IL-6, but not IL-8 nor TNF-alpha, inhibited the cleavage of ULVWF strings by ADAMTS13 under flowing, but not static, conditions. These results suggest that inflammatory cytokines may stimulate the ULVWF release (IL-8 and TNF-alpha) and inhibit the ULVWF cleavage (IL-6), resulting in the accumulation of hyperreactive ULVWF in plasma and on the surface of endothelial cells to induce platelet aggregation and adhesion on the vascular endothelium. The findings describe a potential linkage between inflammation and thrombosis that may be of therapeutic importance.
Collapse
Affiliation(s)
- Aubrey Bernardo
- Thrombosis Research Section, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
27
|
Visseren FL, Verkerk MS, Bouter KP, Diepersloot RJ, Erkelens DW. Interleukin-6 production by endothelial cells after infection with influenza virus and cytomegalovirus. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 1999; 134:623-30. [PMID: 10595791 DOI: 10.1016/s0022-2143(99)90103-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Inflammation plays a role in the pathogenesis of cardiovascular diseases. Viruses may be a cause of chronic inflammation, and both influenza virus and CMV have been associated with cardiovascular diseases. IL-6, a proinflammatory cytokine with antiviral effects, has a pivotal role in the immune response, and under pathologic conditions, prohemostatic effects of IL-6 could lead to pathologic thrombosis and vascular plaque instability. To investigate this role of IL-6, we measured the production of IL-6 by human endothelial cells after infection with influenza virus and CMV. After infection with influenza virus or CMV, IL-6 release into the medium increased (1756.5+/-156.9 pg/mL vs 284.4+/-55.3 pg/mL; P < .001) for influenza-Infected compared with uninfected cells after 36 hours' incubation. Ultracentrifuged influenza virus supernatants, heat-inactivated virus, and purified hemagglutinin were not able to elicit IL-6 synthesis by human endothelial cells. These findings show that CMV and influenza virus are capable of modulating the in vitro production of IL-6, a cytokine involved in vascular inflammation, by human endothelial cells.
Collapse
Affiliation(s)
- F L Visseren
- University Hospital Utrecht, Department of Internal and Vascular Medicine, The Netherlands
| | | | | | | | | |
Collapse
|
28
|
Xie P, Chan FS, Ip NY, Leung MF. Induction of gp130 and LIF by differentiation inducers in human myeloid leukemia K562 cells. Leuk Res 1999; 23:1113-9. [PMID: 10613356 DOI: 10.1016/s0145-2126(99)00141-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
It has been previously shown that phorbol 12-myristate 13-acetate (PMA), a potent differentiation inducer, induced the expression of both interleukin-6 (IL-6) and IL-6 receptor alpha component (IL-6Ralpha) in K562 leukemia cells. In the present study, we examined the ability of several differentiation inducers to regulate the expression of the signal-transducing receptor component for IL-6, gp130, and cytokine leukemia inhibitory factor (LIF) in K562 cells. We found that the expression of gp130 was dramatically induced at both the mRNA and protein levels by the two megakaryocytic inducers sodium butyrate (NaBut) and PMA. In contrast, the mRNA expression of LIF was induced by the two erythroid inducers 1-beta-D-arabinofuranosyl cytosine (Ara-C) and hemin. Furthermore, activation of the PMA-induced gp130 receptor by exogenous IL-6 potentiated the differentiating effects of PMA. Our findings suggest that IL-6/gp130 signaling may be involved in the regulation of the megakaryocytic differentiation of K562 cells.
Collapse
Affiliation(s)
- P Xie
- Department of Biology, Biotechnology Research Institute, The Hong Kong University of Science and Technology, People's Republic of China
| | | | | | | |
Collapse
|
29
|
Furuhashi M, Miyabe Y, Oda H. A case of thrombopoietin-producing ovarian carcinoma confirmed by immunohistochemistry. Gynecol Oncol 1999; 74:278-81. [PMID: 10419745 DOI: 10.1006/gyno.1999.5428] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A case of ovarian carcinoma with thrombopoietin production is reported. A 49-year-old Japanese woman had serous cystadenocarcinoma of the right ovary with peritoneal spread. The platelet count was elevated to 432 x 10(9)/L. Enzyme-linked immunosorbent assay of the serum demonstrated remarkably high levels of thrombopoietin (2.96 fmol/ml). Immunohistochemical examination using an antibody specific for thrombopoietin revealed positive staining in the carcinoma cells, confirming thrombopoietin production.
Collapse
Affiliation(s)
- M Furuhashi
- Department of Obstetrics and Gynecology, Handa City Hospital, Handa, 475-8599, Japan
| | | | | |
Collapse
|
30
|
Funakoshi Y, Ichiki T, Ito K, Takeshita A. Induction of interleukin-6 expression by angiotensin II in rat vascular smooth muscle cells. Hypertension 1999; 34:118-25. [PMID: 10406834 DOI: 10.1161/01.hyp.34.1.118] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent studies suggest that atherosclerosis is a kind of inflammatory process and that cytokine plays important roles in this process. Although it is generally accepted that angiotensin II (Ang II) plays an important role in atherogenesis, the role of Ang II in cytokine production has not been explored. In this report, we investigated the effect of Ang II on the production of interleukin-6 (IL-6), which is a multifunctional proinflammatory cytokine in rat vascular smooth muscle cells. Ang II significantly increased the expression of IL-6 mRNA and protein in a dose-dependent manner (10(-10) to 10(-6) mol/L). The expression of IL-6 mRNA induced by Ang II showed 2 peaks at 30 minutes and 12 to 24 hours after stimulation. The effect of Ang II on IL-6 release and mRNA expression was completely blocked by an Ang II type 1 receptor antagonist, CV11974; however, an Ang II type 2 receptor antagonist, PD123319, showed no effect. Chelating of intracellular Ca(2+) with BAPTA-AM, inhibition of tyrosine kinase with genistein, and inhibition of mitogen-activated protein kinase kinase with PD98059 completely abolished the effect of Ang II. However, downregulation of protein kinase C by pretreatment with a phorbol ester for 24 hours or a specific protein kinase C inhibitor, calphostin C, did not affect the Ang II-induced expression of IL-6 mRNA. Deletion and mutational analysis of IL-6 gene promoter showed that cAMP-responsive element was important for Ang II-induced IL-6 gene expression. Gel mobility shift assay showed an increase of cAMP-responsive element binding protein by Ang II. These results provide new insights into Ang II signaling and the role of Ang II in the progression of inflammatory changes of blood vessels.
Collapse
Affiliation(s)
- Y Funakoshi
- Research Institute of Angiocardiology and Cardiovascular Clinic, Kyushu University School of Medicine, Fukuoka, Japan
| | | | | | | |
Collapse
|
31
|
Russell KE, Perkins PC, Hoffman MR, Miller RT, Walker KM, Fuller FJ, Sellon DC. Platelets from thrombocytopenic ponies acutely infected with equine infectious anemia virus are activated in vivo and hypofunctional. Virology 1999; 259:7-19. [PMID: 10364485 DOI: 10.1006/viro.1999.9737] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Thrombocytopenia is a consistent finding and one of the earliest hematological abnormalities in horses acutely infected with equine infectious anemia virus (EIAV), a lentivirus closely related to human immunodeficiency virus. Multifactorial mechanisms, including immune-mediated platelet destruction and impaired platelet production, are implicated in the pathogenesis of EIAV-associated thrombocytopenia. This study was undertaken to investigate whether regenerative thrombopoiesis and platelet destruction occurred in ponies acutely infected with EIAV. Circulating large, immature platelets were increased in ponies acutely infected with EIAV late in the infection when platelet count was at a nadir. Morphometric analysis of bone marrow from acutely infected ponies revealed significant increased in megakaryocyte area and megakaryocyte nuclear area. A trend toward increased numbers of megakaryocytes was also observed. Platelets from acutely infected ponies had increased surface-bound fibrinogen and ultrastructural changes consistent with in vivo platelet activation. Platelets also had hypofunctional aggregation responses to three agonists in vitro. We conclude that thrombocytopenia in ponies acutely infected with EIAV is regenerative and suggest that bone marrow platelet production is not severely compromised in these ponies. Our findings reveal that in vivo platelet activation occurs in ponies acutely infected with EIAV, and as a result platelets are hypofunctional in vitro. Activation of platelets in vivo may cause platelet degranulation or formation of platelet aggregates, which would result in removal of these damages platelets from circulation. This may represent a form of nonimmune-mediated platelet destruction in ponies acutely infected with EIAV.
Collapse
Affiliation(s)
- K E Russell
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina 27606, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Kanabrocki EL, Sothern RB, Messmore HL, Roitman-Johnson B, McCormick JB, Dawson S, Bremner FW, Third JL, Nemchausky BA, Shirazi P, Scheving LE. Circadian interrelationships among levels of plasma fibrinogen, blood platelets, and serum interleukin-6. Clin Appl Thromb Hemost 1999; 5:37-42. [PMID: 10725981 DOI: 10.1177/107602969900500108] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Circadian (24 h) rhythms of fibrinogen, interleukin-6 (IL-6), and platelet levels were studied in 11 males ages 46 to 72 years. Since there is a known circadian rhythm for fibrinogen and IL-6, we postulated that the peak level (acrophase) of fibrinogen would follow the acrophase of IL-6, based on the fact that IL-6 is the stimulus for fibrinogen production in the liver. Platelet levels were measured to show whether there was any correlation with the IL-6 acrophase because it has been reported that IL-6 affects megakaryocytes and platelets in dogs. We found that the acrophase for IL-6 occurred at 02:03 h and the acrophase for fibrinogen occurred at 09:16 h. Platelet counts peaked at 16:56 h. Thus, there was a positive correlation between IL-6 and fibrinogen acrophases and a negative correlation of each with the acrophase for platelets. The positive linkage of IL-6 with fibrinogen in this study suggests that suppression of IL-6 production would lower those peak fibrinogen levels that occur in the morning in association with arterial ischemic events. This could result in fewer arterial ischemic events, especially in the morning.
Collapse
Affiliation(s)
- E L Kanabrocki
- Department of Nuclear Medicine Service, Veterans Affairs Hospital, Hines, Illinois 60141, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kruth SA. Biological response modifiers: interferons, interleukins, recombinant products, liposomal products. Vet Clin North Am Small Anim Pract 1998; 28:269-95. [PMID: 9556849 DOI: 10.1016/s0195-5616(98)82005-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The concept of enhancing the normal immune response against infections and neoplasms has been considered for decades. The administration of various natural and synthetic products to simulate systemic infections has largely given over to the idea that specific cytokines can be used effectively when administered systemically. Interferons, interleukins, and hematopoietic growth factors may offer substantial clinical benefit in chronic viral infections, and cancers such as osteosarcoma, melanoma, and lymphosarcoma. Erythropoietin has been shown to have great utility in the management of chronic renal failure. At this point in time, only recombinant products derived from humans are commercially available, and they are expensive and not licensed for use in companion animals. Nevertheless, these products may have significant clinical impact on several highly fatal disorders of dogs and cats. When administered systemically, cytokines perturb complex regulatory pathways, and serious side effects may occur. Innovative delivery methods, such as liposomes, gene therapy, and even oral administration may increase the therapeutic index of these molecules. Biological response modification, cytokine biology, and associated delivery systems are rapidly changing fields, and the small animal veterinarian will need to watch for significant advances in these areas over the next several years.
Collapse
Affiliation(s)
- S A Kruth
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Canada
| |
Collapse
|
34
|
Cazenave JP, Gachet C. Anti-platelet drugs: do they affect megakaryocytes? BAILLIERE'S CLINICAL HAEMATOLOGY 1997; 10:163-80. [PMID: 9154321 DOI: 10.1016/s0950-3536(97)80056-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Anti-platelet drugs are used in clinical medicine to prevent thromboembolic complications of cardiovascular diseases. Among anti-platelet drugs, very little is known of their possible effects on megakaryocytes. ASA is the only compound for which it has clearly been demonstrated that its mechanism of action involves acetylation of the Ser 529 residue in cyclo-oxygenase in platelets and megakaryocytes. Because megakaryocytes possess membrane receptors for ADP, the thienopyridine metabolites of ticlopidine and clopidogrel may modify these receptors as in platelets and hence prevent ADP binding and further activation. Megakaryocytes also have GPIIb-IIIa receptors for the adhesive protein fibrinogen and may be accessible in vivo to GPIIb-IIIa antagonists such as the monoclonal antibody abciximab. Drugs such as heparin or the phosphodiesterase inhibitor anagrelide can either inhibit or stimulate megakaryocytopoiesis and platelet production, while cytokines such as thrombopoietin affect megakaryocytopoiesis, platelet production and platelet function by potentiating the activation of platelets by other agonists.
Collapse
|