1
|
Thompson J, Ruan W, Fishman DS, Giefer M, Kim KM, Martinez M, Dall'Oglio L, Balassone V, Torroni F, De Angelis P, Faraci S, Tsai C, Wilsey M, Khalaf R, Mamula P, Liu Q, Zheng Y, Barth BA, Troendle DM. Risk scores for choledocholithiasis perform poorly in patients with hemolytic diseases: a PEDI database report. Front Pediatr 2025; 13:1574462. [PMID: 40264464 PMCID: PMC12011750 DOI: 10.3389/fped.2025.1574462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
Patients with hemolytic diseases are at increased risk for gallstone-related complications. Modified scoring systems have been developed to assess which pediatric patients would benefit from endoscopic retrograde cholangiopancreatography (ERCP) to treat choledocholithiasis. This study aimed to evaluate the ability of the available criteria to determine which pediatric patients with hemolytic diseases are likely to benefit from ERCP. A secondary analysis was performed using the Pediatric ERCP Database Initiative database, which contains prospectively collected data from 1,124 ERCPs at tertiary-care institutions. We compared patients with a hemolytic disease to those without. Data was analyzed by two-tailed Fisher's exact test and paired student t-test. Of the 47 (17.0%) patients who had a hemolytic disease, 34 (72.3%) had one or more common bile duct (CBD) stones at the time of ERCP. Among patients with hemolytic diseases, there were no differences in pre-ERCP imaging or laboratory findings between those with a CBD stone removed at ERCP and those without. Patients with hemolytic diseases did not fit the current choledocholithiasis selection criteria well: 80% in the no-stone at ERCP group met the American Society of Gastrointestinal Endoscopy high-risk criteria, and 90% met the 2016 modified Baylor pediatric criteria. Although not statistically significant, there was an increased number of adverse events in patients with hemolytic diseases. Existing ERCP criteria perform poorly in patients with hemolytic diseases, overestimating their risk of choledocholithiasis. Peri-procedure evaluations such as endoscopic ultrasound, magnetic resonance cholangiopancreatography, and intraoperative cholangiography appear underutilized and may be essential modalities in this population.
Collapse
Affiliation(s)
- Jennifer Thompson
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Douglas S. Fishman
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Matthew Giefer
- Department of Pediatrics, Ochsner Hospital for Children, New Orleans, LA, United States
| | - Kyung Mo Kim
- College of Medicine, University of Ulsan, Ulsan, Republic of Korea
| | - Mercedes Martinez
- Department of Pediatrics, Columbia University, New York City, NY, United States
| | - Luigi Dall'Oglio
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children’s Hospital (IRCCS), Rome, Italy
| | - Valerio Balassone
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children’s Hospital (IRCCS), Rome, Italy
| | - Filippo Torroni
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children’s Hospital (IRCCS), Rome, Italy
| | - Paola De Angelis
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children’s Hospital (IRCCS), Rome, Italy
| | - Simona Faraci
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children’s Hospital (IRCCS), Rome, Italy
| | - Cynthia Tsai
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Michael Wilsey
- Department of Pediatrics, Johns Hopkins All Children’s Hospital, Saint Petersburg, FL, United States
| | - Racha Khalaf
- Department of Pediatrics, University of South Florida, Tampa, FL, United States
| | - Petar Mamula
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Quin Liu
- Department of Pediatrics, Cedars Sinai Medical Center, Los Angeles, CA, United States
| | - Yuhua Zheng
- Department of Pediatrics, University of Southern California, Los Angeles, CA, United States
| | - Bradley A. Barth
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pediatrics, Children’s Health, Children’s Medical Center Dallas, Dallas, TX, United States
| | - David Michael Troendle
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pediatrics, Children’s Health, Children’s Medical Center Dallas, Dallas, TX, United States
| |
Collapse
|
2
|
Qiu Y, Lin J, Wang A, Fang Z, Sakurai Y, Choi H, Williams EK, Hardy ET, Maher K, Coskun AF, Woods G, Lam WA. Clinically relevant clot resolution via a thromboinflammation-on-a-chip. Nature 2025:10.1038/s41586-025-08804-7. [PMID: 40175551 DOI: 10.1038/s41586-025-08804-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/18/2025] [Indexed: 04/04/2025]
Abstract
Thromboinflammation occurs in various diseases, leading to life-threatening microvascular occlusion with resulting end-organ failure1-4. Importantly, how microvascular thromboinflammation resolves remains poorly understood due to the small size-scale of microvasculature and the long duration (weeks to months) of this process. Here we introduce a hydrogel-based thromboinflammation-on-a-chip model with long-term culture capabilities to model microvascular thromboinflammation and monitor clot resolution over clinically and physiologically relevant timescales (up to months). Using this system, we mapped out the distinct temporal phases of clot resolution in microvascular thromboinflammation. Using multiplexed RNA fluorescence in situ hybridization in combination with our thromboinflammation-on-a-chip model, we observed that inflammation shifts the endothelium fibrinolytic balance to favour thrombosis and pinpointed neutrophil elastase as a double-edged sword that induces clot resolution but also tissue damage. We then investigated the mechanisms of potential therapeutic agents that either prevent microvascular thrombosis or accelerate clot resolution. Specifically, we observed that, in thromboinflammation, (1) early tissue plasminogen activator administration within 3 h directly improves endothelial barrier function; (2) prophylactic defibrotide and enoxaparin suppress microvascular thromboinflammation through endothelium-mediated mechanisms; and (3) combining enoxaparin with crizanlizumab reduces microvascular occlusion and protects endothelial function in sickle cell disease. These data introduce a paradigm in investigating the underlying mechanisms of thromboinflammatory clot resolution and conducting drug discovery thereof.
Collapse
Affiliation(s)
- Yongzhi Qiu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Jessica Lin
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Audrey Wang
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhou Fang
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yumiko Sakurai
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hyoann Choi
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Evelyn K Williams
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Elaissa T Hardy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kristin Maher
- Division of Hematology, Oncology, Bone Marrow Transplant, and Cellular Therapy, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Ahmet F Coskun
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Gary Woods
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Wilbur A Lam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
3
|
Zadeh FJ, Fateh A, Saffari H, Khodadadi M, Eslami Samarian M, Nikoubakht N, Dadgar F, Goodarzi V. The vaso-occlusive pain crisis in sickle cell patients: A focus on pathogenesis. Curr Res Transl Med 2025; 73:103512. [PMID: 40220659 DOI: 10.1016/j.retram.2025.103512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
Vaso-occlusive pain crisis (VOC) is recognized as a prominent complication of sickle cell disease, accompanied by debilitating pain and serious consequences for patients, making it the primary cause of visits to hospital emergency departments. In the etiology of VOC, the intricate interaction of endothelial cells, hypoxia, inflammation, and the coagulation system is pivotal. Hemoglobin S polymerization under hypoxic conditions leads to the formation of rigid and adhesive red blood cells that interact with vascular endothelial cells and other blood cells, causing occlusion and subsequent inflammation. Hemolysis of red blood cells results in anemia and heightened inflammation, whereas oxidative stress and involvement of the coagulation system further complicate matters. In this review, we strive to examine the pathophysiology of VOC from these mentioned aspects by consolidating findings from various studies, as a comprehensive understanding of the causes of VOC is essential for the development of targeted therapeutic interventions and the prevention and management of pain, ultimately improving the quality of life for patients.
Collapse
Affiliation(s)
| | - Azadeh Fateh
- Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamed Saffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammadamin Eslami Samarian
- Student Research Committee, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Universal Scientific Education and Research Network(USERN),Tehran,Iran
| | - Nasim Nikoubakht
- Department of Anesthesiology, Hazrat-e Rasool General Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Dadgar
- Department of Internal Medicine, Lorestan University of Medical Science, Khorramabad, Iran; Student Research Committe, Lorestan University of Medical Science, Khorramabad, Iran
| | - Vahid Goodarzi
- Department of Anesthesiology, Rasoul-Akram Medical Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
4
|
Remy T, Jegard J, Chenouard A, Maminirina P, Liet JM, Couec ML, Joram N, Bourgoin P. Characteristics and outcomes of children and young adults with sickle cell disease supported with extracorporeal membrane oxygenation (ECMO): An updated analysis of the ELSO registry. Artif Organs 2025; 49:508-515. [PMID: 39360897 DOI: 10.1111/aor.14880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 02/25/2025]
Abstract
BACKGROUND Sickle cell disease (SCD) is a global hemoglobinopathy; approximately 300 000 individuals are diagnosed annually. Acute chest syndrome (ACS), a common complication, leads to significant hospitalization and mortality, particularly in cases of severe respiratory distress. ECMO outcomes in this specific population are poorly described. METHODS This retrospective observational study, utilizing data from the Extracorporeal Life Support Organization (ELSO) registry, focuses on children and young adults (<40 years) with SCD undergoing ECMO from 1998 to 2022. RESULTS We observed a growing trend in ECMO cases over the last 15 years, with 210 SCD patients identified in the registry (five neonates, 95 children, 110 adults). ECMO was predominantly initiated for pulmonary support (62%), and most of the primary diagnoses were related to SCD (reported as "SCD" or "acute chest syndrome"). The global survival rate was 55.8% (59% for children and 52.7% for adults). None of the children supported for extracorporeal cardiopulmonary resuscitation survived, and only 2/18 (11%) of adults cannulated for ECPR survived. Complication rates, including acute renal failure (33.8%) neurological events (13%), thrombotic (23.3%), or bleeding events (22.9%) were not noticeably different from reported outcomes in the ELSO registry. CONCLUSION Our findings suggest that ECMO outcomes in SCD patients align with general ECMO trends and may not be limited by suspected unfavorable results in children and young adults. Despite limitations, our study contributes valuable insights into using ECMO in SCD, emphasizing the need for further research and understanding in this underexplored domain.
Collapse
Affiliation(s)
- Tancrède Remy
- Department of Pediatrics, La Réunion University Hospital, Saint Denis de la Réunion, France
| | - Julien Jegard
- Pediatric Intensive Care Unit, Nantes University Hospital, Nantes, France
| | - Alexis Chenouard
- Pediatric Intensive Care Unit, Nantes University Hospital, Nantes, France
| | - Pierre Maminirina
- Department of Pediatric Cardiac Surgery, Nantes University Hospital, Nantes, France
| | - Jean Michel Liet
- Pediatric Intensive Care Unit, Nantes University Hospital, Nantes, France
| | - Marie-Laure Couec
- Department of Pediatrics and Pediatric Hematology, Nantes University Hospital, Nantes, France
| | - Nicolas Joram
- Pediatric Intensive Care Unit, Nantes University Hospital, Nantes, France
| | - Pierre Bourgoin
- Pediatric Intensive Care Unit, Department of Anesthesiology, Nantes University Hospital, Nantes, France
| |
Collapse
|
5
|
Bou-Fakhredin R, Cappellini MD, Taher AT, De Franceschi L. Hypercoagulability in hemoglobinopathies: Decoding the thrombotic threat. Am J Hematol 2025; 100:103-115. [PMID: 39400943 DOI: 10.1002/ajh.27500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/15/2024]
Abstract
Beta (β)-thalassemia and sickle cell disease (SCD) are characterized by a hypercoagulable state, which can significantly influence organ complication and disease severity. While red blood cells (RBCs) and erythroblasts continue to play a central role in the pathogenesis of thrombosis in β-thalassemia and SCD, additional factors such as free heme, inflammatory vasculopathy, splenectomy, among other factors further contribute to the complexity of thrombotic risk. Thus, understanding the role of the numerous factors driving this hypercoagulable state will enable healthcare practitioners to enhance preventive and treatment strategies and develop novel therapies for the future. We herein describe the pathogenesis of thrombosis in patients with β-thalassemia and SCD. We also identify common mechanisms underlying the procoagulant profile of hemoglobinopathies translating into thrombotic events. Finally, we review the currently available prevention and clinical management of thrombosis in these patient populations.
Collapse
Affiliation(s)
- Rayan Bou-Fakhredin
- Department of Clinical Sciences and Community, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, SC Medicina ad Indirizzo Metabolico, Milan, Italy
| | - Maria Domenica Cappellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, SC Medicina ad Indirizzo Metabolico, Milan, Italy
| | - Ali T Taher
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Lucia De Franceschi
- Department of Medicine, University of Verona, and Azienda Ospedaliera Universitaria Verona, Verona, Italy
| |
Collapse
|
6
|
Perrone P, Ortega-Luna R, Manna C, Álvarez-Ribelles Á, Collado-Diaz V. Increased Adhesiveness of Blood Cells Induced by Mercury Chloride: Protective Effect of Hydroxytyrosol. Antioxidants (Basel) 2024; 13:1576. [PMID: 39765902 PMCID: PMC11673208 DOI: 10.3390/antiox13121576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Mercury (Hg) is a highly toxic environmental contaminant that can harm human health, ultimately leading to endothelial dysfunction. Hg toxicity is partially mediated by the exposure of the cell membrane's surface of erythrocytes (RBCs) to phosphatidylserine (PS). In the context of these challenges, hydroxytyrosol, a phenolic compound of olive oil, has the ability to mitigate the toxic effects of Hg. This study aims to analyze the effect of Hg on the adhesion of RBCs and polymorphonuclear cells (PMNs) to the vascular endothelium and the potential protective effect of hydroxytyrosol, as these interactions are crucial in the development of cardiovascular diseases (CVDs). RBCs, PMNs, and human vein endothelial cells (HUVECs) were treated with increasing concentrations of HgCl2 and, in some cases, with hydroxytyrosol, and their adhesion to HUVECs and the expression of adhesion molecules were subsequently analyzed. Our results demonstrate that HgCl2 significantly increases the adhesion of both RBCs (2.72 ± 0.48 S.E.M., p-value < 0.02) and PMNs (11.19 ± 1.96 S.E.M., p-value < 0.05) to HUVECs and that their adhesiveness is significantly reduced following treatment with hydroxytyrosol (RBCs, 1.2 ± 1.18 S.E.M., p-value < 0.02 and PMNs, 4.04 ± 1.35 S.E.M., p-value < 0.06). Interestingly, HgCl2 does not alter the expression of adhesion molecules on either HUVECs or RBCs, suggesting that reduced exposure to PS is a key factor in hydroxytyrosol protection against HgCl2-induced RBC adhesion to the endothelium. On the other hand, HgCl2 induces increased expression of several PMN adhesion molecules (CD11b 215.4 ± 30.83 S.E.M. p-value < 0.01), while hydroxytyrosol inhibits their expression (e.g., CD11b 149 ± 14.35 S.E.M., p-value < 0.03), which would seem to be the mechanism by which hydroxytyrosol restricts PMN-endothelium interactions. These results provide new insights into the molecular mechanisms through which hydroxytyrosol mitigates the harmful effects of Hg on cardiovascular health, highlighting its potential as a therapeutic agent that can reduce the cardiovascular risk related to heavy metal exposure.
Collapse
Affiliation(s)
- Pasquale Perrone
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Raquel Ortega-Luna
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (R.O.-L.); (V.C.-D.)
| | - Caterina Manna
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Ángeles Álvarez-Ribelles
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (R.O.-L.); (V.C.-D.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Victor Collado-Diaz
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (R.O.-L.); (V.C.-D.)
| |
Collapse
|
7
|
d’Humières T, Sadraoui Z, Savale L, Boyer L, Guillet H, Alassaad L, de Luna G, Iles S, Balfanz P, Habibi A, Martino S, Amorouayeche Z, Dang TL, Pham Hung d'Alexandry d'Orengiani AL, Rideau D, Train L, Simon T, Ibrahim C, Messonnier LA, Audureau E, Derbel H, Calvet D, Lellouche N, Derumeaux G, Bartolucci P. Atrial arrhythmia in adults with sickle cell anemia: a missing link toward understanding and preventing strokes. Blood Adv 2024; 8:5625-5638. [PMID: 39083808 PMCID: PMC11550361 DOI: 10.1182/bloodadvances.2024013208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/28/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
ABSTRACT Although patients with homozygous sickle cell anemia (SCA) carry both significant left atrial (LA) remodeling and an increased risk of stroke, the prevalence of atrial arrhythmia (AA) has never been prospectively evaluated. The aim of this study was to identify the prevalence and predictors of atrial arrhythmia in SCA. From 2018 to 2022, consecutive adult patients with SCA were included in the DREPACOEUR prospective registry and referred to the physiology department for cardiac evaluation, including a 24-hour electrocardiogram monitoring (ECG-Holter). The primary endpoint was the occurrence of AA, defined by the presence of excessive supraventricular ectopic activity (ESVEA) on ECG-Holter (ie >720 premature atrial contractions [PACs] or any run ≥ 20 PACs) or any recent history of atrial fibrillation. Overall, 130 patients with SCA (mean age: 45±12 years, 48% of male) were included. AA was found in 34 (26%) patients. Age (52±9 vs. 42±12 years, P=0,002), LA dilation (LAVi, 71±24 vs. 52±14 mL/m², P<0.001) and history of stroke without underlying cerebral vasculopathy (26% vs. 5%, P=0.009, OR=6.6 (95%CI 1.4-30.3]) were independently associated with AA. Age and LAVi correlated with PAC load per 24 hours on ECG-Holter. An age over 47 years or a LAVi >55mL/m² could predict AA with a PPV of 33% and a NPV of 92%. AAs are frequent in middle-aged patients with SCA and increase with age and LA remodeling, leading to a major additional risk factor for ischemic stroke. This study provides arguments and means to early screen for AA and potentially prevent cerebral complications.
Collapse
Affiliation(s)
- Thomas d’Humières
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
- INSERM IMRB U955, Université Paris Est, Créteil, France
- Sickle Cell Referral Center-UMGGR, Plateforme d’expertise Maladies Rares Grand Paris Est, Université Paris Est, FHU SENEC, CHU Henri Mondor Assistance Publique Hôpitaux de Paris, Créteil, France
- Paris Cardiovascular Research Center-PARCC, Inserm, Université Paris Cité, Paris, France
| | - Zineb Sadraoui
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Laurent Savale
- Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Assistance Publique Hôpitaux de Paris; Le Kremlin-Bicêtre, France
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Laurent Boyer
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
- INSERM IMRB U955, Université Paris Est, Créteil, France
| | - Henri Guillet
- Sickle Cell Referral Center-UMGGR, Plateforme d’expertise Maladies Rares Grand Paris Est, Université Paris Est, FHU SENEC, CHU Henri Mondor Assistance Publique Hôpitaux de Paris, Créteil, France
- Department of Internal Medicine, Henri Mondor University Hospital, Université Paris Est/Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Lara Alassaad
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
- INSERM IMRB U955, Université Paris Est, Créteil, France
| | - Gonzalo de Luna
- Sickle Cell Referral Center-UMGGR, Plateforme d’expertise Maladies Rares Grand Paris Est, Université Paris Est, FHU SENEC, CHU Henri Mondor Assistance Publique Hôpitaux de Paris, Créteil, France
- Department of Internal Medicine, Henri Mondor University Hospital, Université Paris Est/Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Sihem Iles
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Paul Balfanz
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
- INSERM IMRB U955, Université Paris Est, Créteil, France
| | - Anoosha Habibi
- Sickle Cell Referral Center-UMGGR, Plateforme d’expertise Maladies Rares Grand Paris Est, Université Paris Est, FHU SENEC, CHU Henri Mondor Assistance Publique Hôpitaux de Paris, Créteil, France
- Department of Internal Medicine, Henri Mondor University Hospital, Université Paris Est/Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Suella Martino
- Sickle Cell Referral Center-UMGGR, Plateforme d’expertise Maladies Rares Grand Paris Est, Université Paris Est, FHU SENEC, CHU Henri Mondor Assistance Publique Hôpitaux de Paris, Créteil, France
- Department of Internal Medicine, Henri Mondor University Hospital, Université Paris Est/Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Ziana Amorouayeche
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Thuy Linh Dang
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Anne Laure Pham Hung d'Alexandry d'Orengiani
- Sickle Cell Referral Center-UMGGR, Plateforme d’expertise Maladies Rares Grand Paris Est, Université Paris Est, FHU SENEC, CHU Henri Mondor Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Dominique Rideau
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Laura Train
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Theo Simon
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Christine Ibrahim
- Paris Cardiovascular Research Center-PARCC, Inserm, Université Paris Cité, Paris, France
| | - Laurent A. Messonnier
- Inter-university Laboratory of Human Movement Sciences EA 7424, Université Savoie Mont Blanc, Chambéry, France
- Institut Universitaire de France, Paris, France
| | - Etienne Audureau
- INSERM IMRB U955, Université Paris Est, Créteil, France
- Biostatistics Department, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Haytham Derbel
- Radiology Department, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
- School of Medicine, Université Paris Est, Créteil, France
| | - David Calvet
- Department of Neurology, GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte-Anne, Université Paris Cité, FHU Neurovasc, INSERM 1266, Paris, France
| | - Nicolas Lellouche
- School of Medicine, Université Paris Est, Créteil, France
- Cardiology Department, Rhythmology unit, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Geneviève Derumeaux
- Physiology Department, FHU SENEC, Henri Mondor Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
- INSERM IMRB U955, Université Paris Est, Créteil, France
| | - Pablo Bartolucci
- Sickle Cell Referral Center-UMGGR, Plateforme d’expertise Maladies Rares Grand Paris Est, Université Paris Est, FHU SENEC, CHU Henri Mondor Assistance Publique Hôpitaux de Paris, Créteil, France
- Department of Internal Medicine, Henri Mondor University Hospital, Université Paris Est/Assistance Publique Hôpitaux de Paris, Créteil, France
| |
Collapse
|
8
|
Poliektov NE, Vuncannon DM, Ha TK, Lindsay MK, Chandrasekaran S. The Association between Sickle Cell Disease and Postpartum Severe Maternal Morbidity. Am J Perinatol 2024; 41:2144-2151. [PMID: 38653453 DOI: 10.1055/s-0044-1786174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
OBJECTIVE To compare the risk of severe maternal morbidity (SMM) from the delivery admission to 42 days' postdischarge among persons with sickle cell disease (SCD) to those without SCD. STUDY DESIGN This retrospective cohort study included deliveries ≥20 weeks' gestation at an urban safety net hospital in Atlanta, GA from 2011 to 2019. The exposure was SCD diagnosis. The outcome was a composite of SMM from the delivery admission to 42 days' postdischarge. SMM indicators as defined by the Centers for Disease Control and Prevention were identified using the International Classification of Diseases, Ninth and Tenth Revisions (ICD-9/10) codes; transfusion of blood products and sickle cell crisis were excluded. RESULTS Of N = 17,354 delivery admissions, n = 92 (0.53%) had SCD. Persons with SCD versus without SCD had an increased risk of composite SMM (15.22 vs. 2.29%, p < 0.001), acute renal failure (6.52 vs. 0.71%, p < 0.001), acute respiratory distress syndrome (4.35 vs. 0.17%, p < 0.001), puerperal cerebrovascular disorders (3.26 vs. 0.10%, p < 0.001), sepsis (4.35 vs. 0.42%, p < 0.01), air and thrombotic embolism (5.43 vs. 0.10%, p < 0.001), and ventilation (2.17 vs. 0.09%, p < 0.01). Ultimately, those with SCD had an approximately 6-fold higher incidence risk ratio of SMM, which remained after adjustment for confounders (adjusted incidence risk ratio [aIRR]: 5.96, 95% confidence interval [CI]: 3.4-9.19, p < 0.001). Persons with SCD in active vaso-occlusive crisis at the delivery admission had an approximately 9-fold higher risk of SMM up to 42 days' postdischarge compared with those with SCD not in crisis at the delivery admission (incidence: 25.71 vs. 8.77%, p < 0.05; aIRR: 8.92, 95% CI: 4.5-10.04, p < 0.05). Among those with SCD, SMM at the delivery admission was primarily related to renal and cerebrovascular events, whereas most postpartum SMM was related to respiratory events or sepsis. CONCLUSION SCD is significantly associated with an increased risk of SMM during the delivery admission and through 42 days' postdischarge. Active crisis at delivery further increases the risk of SMM. KEY POINTS · Sickle cell disease was associated with an approximately 6-fold increased risk of SMM.. · Active vaso-occlusive crisis at delivery was associated with an approximately 9-fold increased risk of SMM.. · 48% of SMM events in persons with SCD occurred postpartum and were respiratory- or sepsis-related..
Collapse
Affiliation(s)
- Natalie E Poliektov
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia
| | - Danielle M Vuncannon
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia
| | - Thoa K Ha
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia
| | - Michael K Lindsay
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia
| | - Suchitra Chandrasekaran
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
9
|
Hamdy M, Shaheen IA, Khallaf M, Selim YMM. Thrombin activatable fibrinolysis inhibitor plasma levels and TAFI Thr325Ile genetic polymorphism in a cohort of Egyptian sickle cell disease patients and impact on disease severity. Pediatr Blood Cancer 2024; 71:e30959. [PMID: 38520679 DOI: 10.1002/pbc.30959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/07/2024] [Accepted: 02/28/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Thrombin is a critical protease modulating thrombosis as well as inflammation, which are one of the main pathophysiological mechanisms in sickle vasculopathy, and its levels were reported to be high in sickle cell disease (SCD). The thrombin-thrombomodulin complex activates the TAFI inhibitor of fibrinolysis, which acts by reducing plasmin affinity for its substrate thus hindering fibrinolysis. OBJECTIVE We aimed to determine the influence of the Thr325Ile single nucleotide polymorphism (SNP) on TAFI antigen levels and potential effects on the severity of SCD in a cohort of Egyptian patients. METHODS Genotyping of Thr325lle polymorphism using Taq-Man SNP genotyping assay and TAFI level measurement using an enzyme-linked immunosorbent assay were performed for 80 SCD patients (45 homozygous HbSS, 16 S/β0 and 19 Sβ+) as well as 80 age- and gender-matched healthy control subjects. RESULTS Plasma TAFI levels were higher in SCD patients with Thr325Ile polymorphism, yet the difference was not statistically significant (p = .204). SCD patients with polymorphic genotypes had a greater number of hospital admissions (p = .03). Ten patients with acute chest syndrome had the homozygous polymorphic genotype (GG), and all patients with pulmonary hypertension had the polymorphic genotype (six were homozygous [GG] and five were heterozygous [GA]). Patients with SCD complicated with pulmonary hypertension showed significantly higher plasma TAFI levels (p = .044). CONCLUSION The analysis of Thr325Ile polymorphisms combined with plasma TAFI levels suggests that the analyzed SNP could influence plasma TAFL levels and SCD disease severity and hospitalization rates, which could be predictors for complex disease.
Collapse
Affiliation(s)
- Mona Hamdy
- Department of Pediatrics, Pediatric Hematology and BMT Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Iman A Shaheen
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Khallaf
- Department of Pediatrics, Pediatric Hematology and BMT Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Yasmeen M M Selim
- Department of Pediatrics, Pediatric Hematology and BMT Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
10
|
Žigon P, Shao B. Editorial: Inflammation, the link between venous and arterial thrombosis. Front Cardiovasc Med 2024; 11:1433858. [PMID: 38873263 PMCID: PMC11171132 DOI: 10.3389/fcvm.2024.1433858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Affiliation(s)
- Polona Žigon
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- FAMNIT, University of Primorska, Koper, Slovenia
| | - Bojing Shao
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| |
Collapse
|
11
|
Chatzidavid S, Flevari P, Tombrou I, Anastasiadis G, Dimopoulou M. Pulmonary Hypertension in Sickle Cell Disease: Novel Findings of Gene Polymorphisms Related to Pathophysiology. Int J Mol Sci 2024; 25:4792. [PMID: 38732015 PMCID: PMC11084253 DOI: 10.3390/ijms25094792] [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: 03/29/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Pulmonary hypertension (PH) is a progressive and potentially fatal complication of sickle cell disease (SCD), affecting 6-10% of adult SCD patients. Various mechanisms and theories have been evaluated to explain the pathophysiology of this disease. However, questions remain, particularly regarding the clinical heterogeneity of the disease in terms of symptoms, complications, and survival. Beyond the classical mechanisms that have been thoroughly investigated and include hemolysis, nitric oxide availability, endothelial disorders, thrombosis, and left heart failure, attention is currently focused on the potential role of genes involved in such processes. Potential candidate genes are investigated through next-generation sequencing, with the transforming growth factor-beta (TGF-β) pathway being the initial target. This field of research may also provide novel targets for pharmacologic agents in the future, as is already the case with idiopathic PH. The collection and processing of data and samples from multiple centers can yield reliable results that will allow a better understanding of SCD-related PH as a part of the disease's clinical spectrum. This review attempts to capture the most recent findings of studies on gene polymorphisms that have been associated with PH in SCD patients.
Collapse
Affiliation(s)
| | | | | | | | - Maria Dimopoulou
- Thalassemia and Sickle Cell Disease Unit, Center of Expertise in Rare Hematological Diseases (Hemoglobinopathies), Laikon General Hospital Member of EuroBlood NET, 16 Sevastoupoleos Str., 11526 Athens, Greece; (S.C.); (P.F.); (I.T.); (G.A.)
| |
Collapse
|
12
|
Hersi K, Ramani GV, Law JY, Sadek AS, Vaidya A, Gladwin MT, Cassady SJ. Diagnosis and management of chronic thromboembolic pulmonary hypertension (CTEPH) in sickle cell disease: A review. Pulm Circ 2024; 14:e12362. [PMID: 38803827 PMCID: PMC11128985 DOI: 10.1002/pul2.12362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/13/2024] [Accepted: 03/11/2024] [Indexed: 05/29/2024] Open
Abstract
Pulmonary hypertension in sickle cell disease (SCD) is a complex phenomenon resulting from multiple overlapping etiologies, including pulmonary vasoconstriction in the setting of chronic hemolytic anemia, diastolic dysfunction, and chronic thromboembolic disease. The presence of pulmonary hypertension of any cause in SCD confers a significant increase in mortality risk. Evidence to guide the management of patients with sickle cell disease and chronic thromboembolic pulmonary hypertension (CTEPH) is scant and largely the realm of case reports and small case series. Centered on a discussion of a complex young patient with hemoglobin hemoglobin SC who ultimately underwent treatment with pulmonary thromboendarterectomy, we review the available literature to guide management and discuss and overview of treatment of CTEPH in SCD, considering the unique considerations and challenges facing patients suffering from this multisystem disease.
Collapse
Affiliation(s)
- Kadija Hersi
- Division of Pulmonary and Critical Care Medicine, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Gautam V. Ramani
- Division of Cardiology, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Jennie Y. Law
- Division of Hematology and Oncology, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Ahmed S. Sadek
- Division of Cardiology, Department of MedicineLewis Katz School of Medicine at TemplePhiladelphiaPennsylvaniaUSA
| | - Anjali Vaidya
- Division of Cardiology, Department of MedicineLewis Katz School of Medicine at TemplePhiladelphiaPennsylvaniaUSA
| | - Mark T. Gladwin
- Division of Pulmonary and Critical Care Medicine, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Steven J. Cassady
- Division of Pulmonary and Critical Care Medicine, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
13
|
Sussman RG, Mburu J, Steele M, Bang A, Friedman J, Goldman R, Kirby M, Rand ML, Blanchette VS, Pluthero FG, Williams S, Kahr WH. Constitutive hypercoagulability in pediatric sickle cell disease patients with hemoglobin SS genotype. Res Pract Thromb Haemost 2024; 8:102374. [PMID: 38605827 PMCID: PMC11004888 DOI: 10.1016/j.rpth.2024.102374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 04/13/2024] Open
Abstract
Background Constitutive inflammation and hemostatic activation have been identified as key contributors to the pathophysiology of sickle cell disease (SCD), leading to clinical consequences such as vaso-occlusive crises and stroke. Patients with hemoglobin SS (HbSS) and hemoglobin SC (HbSC) genotypes are reported to have different symptoms, as do patients in steady-state and crisis situations. Differences among these groups remain unclear in pediatric patients. Objectives To compare hemostatic activity in HbSS and HbSC pediatric patients during steady state, in crisis, and in clinical follow-up and compare HbSS and HbSC patients with normal healthy children. Methods Whole-blood coagulation assay thromboelastography (TEG) was used to assess hemostatic activity. In parallel, flow cytometry was used to assess procoagulant surface expression of platelets and red blood cells. Results TEG results indicated no significant differences in clotting onset (R time), clot maximum amplitude, or maximum rate of thrombus generation among steady-state, crisis, and follow-up subgroups of HbSS and HbSC patients. TEG parameters did not differ significantly between HbSC patients and healthy children, while HbSS patients showed significantly shorter R time and greater maximum amplitude and maximum rate of thrombus generation, all indicative of a constitutive hypercoagulable state. Flow cytometry results did not detect increased platelet integrin αIIbβ3 activation or red blood cell procoagulant surface expression in SCD patients compared with unaffected children. Conclusion Our results indicate that pediatric SCD patients with the HbSS genotype have constitutively activated hemostasis relative to HbSC patients and healthy children. It remains to be determined how treatments that improve clinical outcomes in SCD patients affect this constitutively hypercoagulable state.
Collapse
Affiliation(s)
- Raizl G. Sussman
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joy Mburu
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - MacGregor Steele
- Department of Pediatrics, Section of Pediatric Hematology, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Annie Bang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Jeremy Friedman
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Ran Goldman
- Division of Clinical Pharmacology and Pediatric Emergency Medicine, Department of Pediatrics, British Columbia Children’s Hospital, BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Melanie Kirby
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Margaret L. Rand
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Translational Medicine Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Victor S. Blanchette
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Fred G. Pluthero
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Suzan Williams
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Walter H.A. Kahr
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Davila J, Mitchell WB, Morrone K, Silver EJ, Minniti CP, Billett HH, Desai PC, O'Brien SH, Manwani D. Venous thromboembolism prophylaxis practices for patients with sickle cell disease prior to and during the COVID-19 pandemic. Blood Coagul Fibrinolysis 2023; 34:471-477. [PMID: 37756203 DOI: 10.1097/mbc.0000000000001250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Patients with sickle cell disease (SCD) are predisposed to a hypercoagulable state due to alterations in the coagulation system. Despite concern for the development of venous thromboembolism (VTE) in this population, there are no standardized guidelines for routine thromboprophylaxis. The objective of this study was to assess thromboprophylaxis practices of adult and pediatric treaters of SCD before and during the coronavirus disease of 2019 (COVID-19) pandemic. A cross-sectional electronic survey was distributed to pediatric and adult hematology oncology practitioners through seven SCD-specific interest groups between May 29, 2020, and July 13, 2020. Of 93 total responses, 14% ( N = 13) reported they only treat patients more than 21 years old; 38.7% ( N = 36) only treat patients 0-21 years old and 47.3% ( N = 44) reported they treat both. Our study showed that before the COVID-19 pandemic, 96% of adult practitioners would recommend pharmacologic thromboprophylaxis, mechanical thromboprophylaxis or both for hospitalized adults with thromboprophylaxis, but only 76% of pediatric treaters would recommend any thromboprophylaxis in hospitalized children ( P < 0.0001), with 24% of pediatric treaters choosing no thromboprophylaxis at all. During the COVID-19 pandemic, pharmacologic thromboprophylaxis specifically was recommended for adults by 94% of treaters and for pediatric patients by 76% of treaters. These findings suggest that despite the lack of evidence-based thromboprophylaxis guidelines in adults and children with thromboprophylaxis, subspecialty treaters routinely provide pharmacologic thromboprophylaxis in their adult patients and will modify their practice in pediatric patients who are considered at a high risk for VTE.
Collapse
Affiliation(s)
| | | | | | - Ellen J Silver
- Division of Academic General Pediatrics, Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine
| | - Caterina P Minniti
- Division of Hematology, Departments of Oncology and Medicine, Montefiore Health System and the Albert Einstein College of Medicine, Bronx, New York
| | - Henny H Billett
- Division of Hematology, Departments of Oncology and Medicine, Montefiore Health System and the Albert Einstein College of Medicine, Bronx, New York
| | - Payal C Desai
- Division of Hematology, Atrium Health Levine Cancer Institute, Morehead Medical Drive, Charlotte, North Carolina
| | - Sarah H O'Brien
- Division of Pediatric Hematology/Oncology, Nationwide Children's Hospital/The Ohio State University, Columbus, Ohio, USA
| | | |
Collapse
|
15
|
Fogarty H, Ahmad A, Atiq F, Doherty D, Ward S, Karampini E, Rehill A, Leon G, Byrne C, Geoghegan R, Conroy H, Byrne M, Budde U, Schneppenheim S, Sheehan C, Ngwenya N, Baker RI, Preston RJS, Tuohy E, McMahon C, O’Donnell JS. VWF-ADAMTS13 axis dysfunction in children with sickle cell disease treated with hydroxycarbamide vs blood transfusion. Blood Adv 2023; 7:6974-6989. [PMID: 37773926 PMCID: PMC10690561 DOI: 10.1182/bloodadvances.2023010824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/17/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023] Open
Abstract
Previous studies have reported elevated von Willebrand factor (VWF) levels in patients with sickle cell disease (SCD) and demonstrated a key role for the VWF-ADAMTS13 axis in the pathobiology of SCD vaso-occlusion. Although blood transfusion is the gold standard for stroke prevention in SCD, the biological mechanisms underpinning its improved efficacy compared with hydroxycarbamide are not fully understood. We hypothesized that the improved efficacy of blood transfusion might relate to differences in VWF-ADAMTS13 axis dysfunction. In total, 180 children with a confirmed diagnosis of SCD (hemoglobin SS) on hydroxycarbamide (n = 96) or blood transfusion (n = 84) were included. Despite disease-modifying treatment, plasma VWF and VWF propeptide were elevated in a significant proportion of children with SCD (33% and 47%, respectively). Crucially, all VWF parameters were significantly higher in the hydroxycarbamide compared with the blood transfusion cohort (P < .05). Additionally, increased levels of other Weibel-Palade body-stored proteins, including factor VIII (FVIII), angiopoietin-2, and osteoprotegerin were observed, indicated ongoing endothelial cell activation. Children treated with hydroxycarbamide also had higher FVIII activity and enhanced thrombin generation compared with those in the blood transfusion cohort (P < .001). Finally, hemolysis markers strongly correlated with VWF levels (P < .001) and were significantly reduced in the blood transfusion cohort (P < .001). Cumulatively, to our knowledge, our findings demonstrate for the first time that despite treatment, ongoing dysfunction of the VWF-ADAMTS13 axis is present in a significant subgroup of pediatric patients with SCD, especially those treated with hydroxycarbamide.
Collapse
Affiliation(s)
- Helen Fogarty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Azaz Ahmad
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ferdows Atiq
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Dearbhla Doherty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Soracha Ward
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ellie Karampini
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aisling Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Gemma Leon
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ciara Byrne
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Rosena Geoghegan
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Helena Conroy
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Mary Byrne
- National Coagulation Centre, St. James’s Hospital, Dublin, Ireland
| | - Ulrich Budde
- Department of Haemostaseology, MVZ Medilys Laborgesellschaft mbH, Hamburg, Germany
| | - Sonja Schneppenheim
- Department of Haemostaseology, MVZ Medilys Laborgesellschaft mbH, Hamburg, Germany
| | - Ciara Sheehan
- Department of Haematology, St. James’s Hospital, Dublin, Ireland
| | - Noel Ngwenya
- Department of Haematology, St. James’s Hospital, Dublin, Ireland
| | - Ross I. Baker
- Western Australia Centre for Thrombosis and Haemostasis, Perth Blood Institute, Murdoch University, Perth, WA, Australia
- Irish-Australian Blood Collaborative Network, Dublin, Ireland and Perth, Australia
| | - Roger J. S. Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Emma Tuohy
- Department of Haematology, St. James’s Hospital, Dublin, Ireland
| | - Corrina McMahon
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - James S. O’Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
- National Coagulation Centre, St. James’s Hospital, Dublin, Ireland
- Irish-Australian Blood Collaborative Network, Dublin, Ireland and Perth, Australia
| |
Collapse
|
16
|
Hamali HA. Hypercoagulability in Sickle Cell Disease: A Thrombo-Inflammatory Mechanism. Hemoglobin 2023; 47:205-214. [PMID: 38189099 DOI: 10.1080/03630269.2023.2301026] [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: 04/20/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
Sickle cell disease (SCD) is a group of inherited disorders characterized by the presence of abnormal hemoglobin S. Patients with SCD suffer from frequent episodes of anemia, chronic hemolysis, pain crisis, and vaso-occlusion. Additionally, SCD is associated with diverse and serious clinical complications, including thrombosis, which can lead to organ failure, increased morbidity, and eventually, mortality. SCD is known to be a hypercoagulable condition, and the cause of hypercoagulability is multifactorial, with the molecular basis of hemoglobin S being the main driver. The presence of hemoglobin S induces sickling of the RBCs and their subsequent hemolysis, as well as oxidative stress. Both of these processes can alter the hemostatic system, through the activation of platelets, coagulation system, and fibrinolysis, as well as depletion of coagulation inhibitors. These changes can also induce the formation of microvesicles and expression of tissue factor, leading to activation of WBCs, endothelial cell damage, and inflammatory response. Understanding the various factors that drive hypercoagulability as a thrombo-inflammatory mechanism in SCD can help provide explanations for the pathogenesis and other complications of the disease.
Collapse
Affiliation(s)
- Hassan A Hamali
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| |
Collapse
|
17
|
Alshabeeb MA, Alwadaani D, Al Qahtani FH, Abohelaika S, Alzahrani M, Al Zayed A, Al Saeed HH, Al Ajmi H, Alsomaie B, Rashid M, Daly AK. Impact of Genetic Variations on Thromboembolic Risk in Saudis with Sickle Cell Disease. Genes (Basel) 2023; 14:1919. [PMID: 37895268 PMCID: PMC10606407 DOI: 10.3390/genes14101919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/04/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Sickle cell disease (SCD) is a Mendelian disease characterized by multigenic phenotypes. Previous reports indicated a higher rate of thromboembolic events (TEEs) in SCD patients. A number of candidate polymorphisms in certain genes (e.g., FVL, PRT, and MTHFR) were previously reported as risk factors for TEEs in different clinical conditions. This study aimed to genotype these genes and other loci predicted to underlie TEEs in SCD patients. METHODOLOGY A multi-center genome-wide association study (GWAS) involving Saudi SCD adult patients with a history of TEEs (n = 65) and control patients without TEE history (n = 285) was performed. Genotyping used the 10× Affymetrix Axiom array, which includes 683,030 markers. Fisher's exact test was used to generate p-values of TEE associations with each single-nucleotide polymorphism (SNP). The haplotype analysis software tool version 1.05, designed by the University of Göttingen, Germany, was used to identify the common inherited haplotypes. RESULTS No association was identified between the targeted single-nucleotide polymorphism rs1801133 in MTHFR and TEEs in SCD (p = 0.79). The allele frequency of rs6025 in FVL and rs1799963 in PRT in our cohort was extremely low (<0.01); thus, both variants were excluded from the analysis as no meaningful comparison was possible. In contrast, the GWAS analysis showed novel genome-wide associations (p < 5 × 10-8) with seven signals; five of them were located on Chr 11 (rs35390334, rs331532, rs317777, rs147062602, and rs372091), one SNP on Chr 20 (rs139341092), and another on Chr 9 (rs76076035). The other 34 SNPs located on known genes were also detected at a signal threshold of p < 5 × 10-6. Seven of the identified variants are located in olfactory receptor family 51 genes (OR51B5, OR51V1, OR51A1P, and OR51E2), and five variants were related to family 52 genes (OR52A5, OR52K1, OR52K2, and OR52T1P). The previously reported association between rs5006884-A in OR51B5 and fetal hemoglobin (HbF) levels was confirmed in our study, which showed significantly lower levels of HbF (p = 0.002) and less allele frequency (p = 0.003) in the TEE cases than in the controls. The assessment of the haplotype inheritance pattern involved the top ten significant markers with no LD (rs353988334, rs317777, rs14788626882, rs49188823, rs139349992, rs76076035, rs73395847, rs1368823, rs8888834548, and rs1455957). A haplotype analysis revealed significant associations between two haplotypes (a risk, TT-AA-del-AA-ins-CT-TT-CC-CC-AA, and a reverse protective, CC-GG-ins-GG-del-TT-CC-TT-GG-GG) and TEEs in SCD (p = 0.024, OR = 6.16, CI = 1.34-28.24, and p = 0.019, OR = 0.33, CI = 0.13-0.85, respectively). CONCLUSIONS Seven markers showed novel genome-wide associations; two of them were exonic variants (rs317777 in OLFM5P and rs147062602 in OR51B5), and less significant associations (p < 5 × 10-6) were identified for 34 other variants in known genes with TEEs in SCD. Moreover, two 10-SNP common haplotypes were determined with contradictory effects. Further replication of these findings is needed.
Collapse
Affiliation(s)
- Mohammad A. Alshabeeb
- King Abdullah International Medical Research Center (KAIMRC), Riyadh 11426, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
| | - Deemah Alwadaani
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Riyadh 11481, Saudi Arabia
| | - Farjah H. Al Qahtani
- Hematology/Oncology Center, King Saud University Medical City (KSUMC), Riyadh 11411, Saudi Arabia;
| | - Salah Abohelaika
- Research Department, Qatif Central Hospital (QCH), Qatif 32654, Saudi Arabia;
- Pharmacy Department, Qatif Central Hospital (QCH), Qatif 32654, Saudi Arabia
| | - Mohsen Alzahrani
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
- King Fahad Hospital, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Abdullah Al Zayed
- Hematology Department, Qatif Central Hospital (QCH), Qatif 32654, Saudi Arabia; (A.A.Z.); (H.H.A.S.)
| | - Hussain H. Al Saeed
- Hematology Department, Qatif Central Hospital (QCH), Qatif 32654, Saudi Arabia; (A.A.Z.); (H.H.A.S.)
| | - Hala Al Ajmi
- King Abdullah International Medical Research Center (KAIMRC), Riyadh 11426, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
| | - Barrak Alsomaie
- King Abdullah International Medical Research Center (KAIMRC), Riyadh 11426, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
| | - Mamoon Rashid
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
- Department of AI and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), Riyadh 11481, Saudi Arabia
| | - Ann K. Daly
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| |
Collapse
|
18
|
Khan SA, Halawani SH, Zughaibi TA, Khan SA. Potential inflammatory targets in the integrative health care of patients with sickle cell disease. Exp Ther Med 2023; 26:485. [PMID: 37745045 PMCID: PMC10515103 DOI: 10.3892/etm.2023.12184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/02/2023] [Indexed: 09/26/2023] Open
Abstract
Inflammation plays an integral role in the complications of sickle cell disease (SCD), which can lead to vaso-occlusive crisis and extreme pain. SCD is accompanied by numerous complications, including cardiovascular disease, cognitive decline and endothelial dysfunction, contributing to mortality. As disease severity increases with age, the present study aimed to assess if age is also correlated with a definite pattern of progression of the two inflammatory markers, high-sensitivity C-reactive protein (hsCRP) and total homocysteine (tHCY). The findings of the present study could lead to an improved understanding of the threshold levels of these inflammatory markers and timely interventions to delay complications. In an observational study, levels of hsCRP and tHCY were analyzed in 70 patients (35 male and 35 female patients) with SCD aged between 5 and 16 years. hsCRP levels were in the high-risk range in 64.29% (n=45) of all male and female patients. A sex-wise distribution showed that, of the 35 male patients, 74.28% (n=26) were in the high-risk range, and of the 35 female patients, 54.28% (n=19) were in the high-risk range. An age-wise distribution showed that of the 41 patients in the 5-10-years age group, 70.73% (n=29), were in the high-risk range. In comparison, of the 29 patients in the 11-16-years age group, 55.17% (n=16) were in the high-risk range. tHCY levels were observed to be in the normal range in 98.57% (n=69) of all children, as compared with 1.43% (n=1) in the high-risk range. Furthermore, a sex-wise distribution showed that female patients in the high-risk group of hsCRP had higher concentrations of tHCY as compared with the male patients in that risk group. An age-wise distribution of hsCRP concentration also showed that the risk of CVD in patients in the 11-16-years age group was higher with increased concentrations of tHCY. A weak negative correlation was observed between age and hsCRP concentrations (r-value=-0.280; P=0.026) and a weak positive correlation was detected between tHCY and age (r-value=0.259; P=0.036). In conclusion, the results of the present study indicated that higher levels of hsCRP could be a useful marker in children with SCD, and levels of tHCY may be an adjunct marker as the disease progresses with age.
Collapse
Affiliation(s)
- Shahida A Khan
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Saeed H Halawani
- Department of Hematology and Immunology, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Torki Al Zughaibi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Sarah A Khan
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
19
|
Hashemi Tayer A, Ranjbaran R, Kamravan M, Abbasi M, Zareian R. Association of Circulating Procoagulant Microvesicles with Painful Vaso-Occlusive Crisis in Sickle Cell Disease. Transfus Med Hemother 2023; 50:448-455. [PMID: 37936632 PMCID: PMC10626395 DOI: 10.1159/000525640] [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: 10/01/2021] [Accepted: 06/20/2022] [Indexed: 11/09/2023] Open
Abstract
Introduction Thrombotic complication is one of the features of sickle cell disease (SCD), characterized by appearance of phosphatidylserine on the outer membrane of sickle-shaped red blood cells and most abundantly on membrane protrusions called microvesicles (MVs). However, the exact mechanism by which MVs may enhance coagulant activity in SCD patients has not been fully addressed. The aim of this study was to further investigate the procoagulant activity of circulating MVs in sickle cell crises. Materials and Methods Subjects included in this cross-sectional study were 47 patients with SCD and 25 normal subjects with written informed consent obtained from all the participants. MV analysis was conducted by using CD61, CD235α, and Annexin-V monoclonal antibodies. The coagulant activity of MVs was determined by an ELISA-based procoagulant activity assay. Results The majority of MVs were originated from platelets (CD61+) and erythrocytes (CD235+). These MVs demonstrated significantly enhanced levels during the painful crisis when compared with the steady-state period (p < 0.001) and controls (p < 0.001). Also, the procoagulant activity of MVs was significantly higher in crisis compared to those of steady state (p < 0.001) and positively correlated with the number of Annexin-V+ MVs (p < 0.001). Significant correlations were found between erythrocyte-derived MVs with hemolysis marker (r = 0.51, p < 0.001) and the hemoglobin level (r = -0.63, p < 0.001). Conclusion The numbers of platelet- and erythrocyte-derived MVs are related to painful crisis, and their quantification in SCD may be helpful for identifying cases at increased risk of thrombotic complications.
Collapse
Affiliation(s)
- Akbar Hashemi Tayer
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Reza Ranjbaran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Kamravan
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mojdeh Abbasi
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Reyhaneh Zareian
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| |
Collapse
|
20
|
Abdi SS, De Haan M, Kirkham FJ. Neuroimaging and Cognitive Function in Sickle Cell Disease: A Systematic Review. CHILDREN 2023; 10:children10030532. [PMID: 36980090 PMCID: PMC10047189 DOI: 10.3390/children10030532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 03/12/2023]
Abstract
Sickle cell disease (SCD) is the most common inherited single-gene disease. Complications include chronic anaemia, reduced oxygen-carrying capability, and cerebral vasculopathy, resulting in silent cerebral infarction, stroke, and cognitive dysfunction with impairments in measures of executive function, attention, reasoning, language, memory, and IQ. This systematic review aims to investigate the association between neuroimaging findings and cognition in children with SCD. Searches of PubMed and Embase were conducted in March 2022. Studies were included if participants were <18 years, if original data were published in English between 1960 and 2022, if any genotype of SCD was included, and if the relationship between cognition and neuroimaging was examined. Exclusion criteria included case studies, editorials, and reviews. Quality was assessed using the Critical Appraisal Skills Programme Case Control Checklist. A total of 303 articles were retrieved; 33 met the eligibility criteria. The presence of overt or silent strokes, elevated blood flow velocities, abnormal functional connectivity, and decreased fMRI activation were associated with neuropsychological deficits in children with SCD when compared to controls. There is a critical need to address the disease manifestations of SCD early, as damage appears to begin at a young age. Most studies were cross-sectional, restricting the interpretation of the directionality of relationships. Future research employing longitudinal neuroimaging and neuropsychological assessments could improve our understanding of the cumulative consequences of SCD on the developing brain.
Collapse
Affiliation(s)
- Suad S. Abdi
- Developmental Neurosciences Section, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Michelle De Haan
- Developmental Neurosciences Section, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Fenella J. Kirkham
- Developmental Neurosciences Section, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Clinical and Experimental Sciences, University of Southampton, Southampton SO16 6YD, UK
- Child Health, University Hospital Southampton, Southampton SO16 6YD, UK
- Correspondence:
| |
Collapse
|
21
|
Chiang KC, Gupta A, Sundd P, Krishnamurti L. Thrombo-Inflammation in COVID-19 and Sickle Cell Disease: Two Faces of the Same Coin. Biomedicines 2023; 11:338. [PMID: 36830874 PMCID: PMC9953430 DOI: 10.3390/biomedicines11020338] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/26/2023] Open
Abstract
People with sickle cell disease (SCD) are at greater risk of severe illness and death from respiratory infections, including COVID-19, than people without SCD (Centers for Disease Control and Prevention, USA). Vaso-occlusive crises (VOC) in SCD and severe SARS-CoV-2 infection are both characterized by thrombo-inflammation mediated by endothelial injury, complement activation, inflammatory lipid storm, platelet activation, platelet-leukocyte adhesion, and activation of the coagulation cascade. Notably, lipid mediators, including thromboxane A2, significantly increase in severe COVID-19 and SCD. In addition, the release of thromboxane A2 from endothelial cells and macrophages stimulates platelets to release microvesicles, which are harbingers of multicellular adhesion and thrombo-inflammation. Currently, there are limited therapeutic strategies targeting platelet-neutrophil activation and thrombo-inflammation in either SCD or COVID-19 during acute crisis. However, due to many similarities between the pathobiology of thrombo-inflammation in SCD and COVID-19, therapies targeting one disease may likely be effective in the other. Therefore, the preclinical and clinical research spurred by the COVID-19 pandemic, including clinical trials of anti-thrombotic agents, are potentially applicable to VOC. Here, we first outline the parallels between SCD and COVID-19; second, review the role of lipid mediators in the pathogenesis of these diseases; and lastly, examine the therapeutic targets and potential treatments for the two diseases.
Collapse
Affiliation(s)
| | - Ajay Gupta
- KARE Biosciences, Orange, CA 89128, USA
- Division of Nephrology, Hypertension and Kidney Transplantation, Department of Medicine, University of California Irvine (UCI) School of Medicine, Irvine, CA 92868, USA
| | - Prithu Sundd
- Vascular Medicine Institute and Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Lakshmanan Krishnamurti
- Division of Pediatric Hematology-Oncology, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
22
|
Mustafa AEM, Tahir NM, Ahmed Mohamed NAE, Mohammed AA, Mohammed SI. Deep Vein Thrombosis of the Left Lower Limb in a Sudanese Child with Sickle Cell Disease. MEDICINES (BASEL, SWITZERLAND) 2022; 9:52. [PMID: 36355057 PMCID: PMC9697196 DOI: 10.3390/medicines9110052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
This is a case of an eleven-year-old female Sudanese child, a known Sickle Cell Anemia (SCA) patient, who presented with fever, as well as left thigh and leg swelling that was associated with pain and warmness, which was diagnosed as Deep Vein Thrombosis (DVT) of her left lower limb. She had a previous history of admissions to the emergency room, during which she once received blood. The patient was managed by carrying out a basic routine initial laboratory investigation. A Doppler ultrasound scan showed features consistent with DVT. Based on the clinical findings and investigation results, management began by providing the patient with intravenous fluid, analgesia, packed Red Blood Cells (RBCs), intravenous antibiotics, and low-molecular-weight heparin. Further consultations showed that there was no need for vascular surgery or surgical intervention. This case highlights the need for more studies on DVT and Venous Thromboembolism (VTE) complications in children with SCA, so as to develop strategies for diagnosis and management in order to reduce the risk of life-threatening complications of VTE in patients with Sickle Cell Disease SCD.
Collapse
Affiliation(s)
- Alam Eldin Musa Mustafa
- Department of Child Health, College of Medicine, King Khalid University, P.O. Box 641, Abha 61421, Saudi Arabia
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Kordofan University, P.O. Box 160, El Obeid 51111, Sudan
| | - Niemat Mohammed Tahir
- Department of Child Health, College of Medicine, King Khalid University, P.O. Box 641, Abha 61421, Saudi Arabia
| | | | | | | |
Collapse
|
23
|
Mubeen S, Domingo-Fernández D, Díaz del Ser S, Solanki DM, Kodamullil AT, Hofmann-Apitius M, Hopp MT, Imhof D. Exploring the Complex Network of Heme-Triggered Effects on the Blood Coagulation System. J Clin Med 2022; 11:jcm11195975. [PMID: 36233841 PMCID: PMC9572022 DOI: 10.3390/jcm11195975] [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: 09/20/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/16/2022] Open
Abstract
Excess labile heme, occurring under hemolytic conditions, displays a versatile modulator in the blood coagulation system. As such, heme provokes prothrombotic states, either by binding to plasma proteins or through interaction with participating cell types. However, despite several independent reports on these effects, apparently contradictory observations and significant knowledge gaps characterize this relationship, which hampers a complete understanding of heme-driven coagulopathies and the development of suitable and specific treatment options. Thus, the computational exploration of the complex network of heme-triggered effects in the blood coagulation system is presented herein. Combining hemostasis- and heme-specific terminology, the knowledge available thus far was curated and modeled in a mechanistic interactome. Further, these data were incorporated in the earlier established heme knowledge graph, "HemeKG", to better comprehend the knowledge surrounding heme biology. Finally, a pathway enrichment analysis of these data provided deep insights into so far unknown links and novel experimental targets within the blood coagulation cascade and platelet activation pathways for further investigation of the prothrombotic nature of heme. In summary, this study allows, for the first time, a detailed network analysis of the effects of heme in the blood coagulation system.
Collapse
Affiliation(s)
- Sarah Mubeen
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
- Enveda Biosciences, Inc., San Francisco, CA 94080, USA
| | - Sara Díaz del Ser
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
- Polytechnic University of Madrid, E-28040 Madrid, Spain
| | - Dhwani M. Solanki
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Alpha T. Kodamullil
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
- Causality Biomodels, Kinfra Hi-Tech Park, Kalamassery, Cochin 683503, Kerala, India
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
| | - Marie-T. Hopp
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Correspondence: (M.-T.H.); (D.I.); Tel.: +49-228-73-5231 (M.-T.H.); +49-228-73-5254 (D.I.)
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Correspondence: (M.-T.H.); (D.I.); Tel.: +49-228-73-5231 (M.-T.H.); +49-228-73-5254 (D.I.)
| |
Collapse
|
24
|
Feugray G, Kasonga F, Grall M, Dumesnil C, Benhamou Y, Brunel V, Le Cam Duchez V, Lahary A, Billoir P. Investigation of thrombin generation assay to predict vaso-occlusive crisis in adulthood with sickle cell disease. Front Cardiovasc Med 2022; 9:883812. [PMID: 36277754 PMCID: PMC9579298 DOI: 10.3389/fcvm.2022.883812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Sickle cell disease (SCD) is an inherited hemoglobinopathy disorder. The main consequence is synthesis of hemoglobin S leading to chronic hemolysis associated with morbidity. The aim of this study was to investigate Thrombin Generation Assay (TGA) to assess hypercoagulability in SCD and TGA parameters as biomarkers of vaso-occlusive crisis (VOC) risk and hospitalization within 1 year. Materials and methods We performed TGA in platelet poor plasma (PPP) with 1 pM of tissue factor and 4 μM of phospholipid-standardized concentration, in duplicate for patients and controls. We measured thrombomodulin (TM), soluble endothelial Protein C Receptor and Tissue Factor Pathway Inhibitor (TFPI). Results A total of 113 adult patients with SCD, 83 at steady state and 30 during VOC, and 25 healthy controls matched on age and gender were included. Among the 83 patients at steady state, (36 S/S-1 S/β0, 20 S/Sα3.7, and 19 S/C-7 S/β+) 28 developed a VOC within 1 year (median: 4 months [2.25–6]). We observed an increase of peak and velocity associated with a shortening of lagtime and time to peak (TTP) and no difference of endogenous thrombin potential (ETP) in patients compared to controls. TFPI (p < 0.001) and TM (p = 0.006) were significantly decreased. TGA confirmed hypercoagulability in all SCD genotypes and clinical status. The association of ETP > 1,207 nM.min and peak >228.5 nM presented a sensitivity of 73.5% and a specificity of 93.9% to predict VOC development within 1 year. Conclusion We have demonstrated a hypercoagulable state in SCD associated with chronic hemolysis. These preliminary findings suggest that TGA parameters, as ETP and peak, could be used to predict VOC development within 1 year.
Collapse
Affiliation(s)
- Guillaume Feugray
- Vascular Hemostasis Unit, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France
| | | | | | - Cécile Dumesnil
- Department of Pediatric Onco-Hematology, CHU Rouen, Rouen, France
| | - Ygal Benhamou
- Department of Internal Medicine, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France
| | - Valery Brunel
- Department of General Biochemistry, CHU Rouen, Rouen, France
| | - Véronique Le Cam Duchez
- Vascular Hemostasis Unit, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France
| | | | - Paul Billoir
- Vascular Hemostasis Unit, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France,*Correspondence: Paul Billoir, ; orcid.org/0000-0001-5632-7713
| |
Collapse
|
25
|
Saade EA, Hojat LS, Gundelly P, Salata RA. Prevention and treatment of COVID-19 in patients with benign and malignant blood disorders. Best Pract Res Clin Haematol 2022; 35:101375. [PMID: 36494144 PMCID: PMC9398935 DOI: 10.1016/j.beha.2022.101375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/27/2022] [Accepted: 08/11/2022] [Indexed: 12/15/2022]
Abstract
Patients with moderate to severe immunosuppression, a condition that is common in many hematologic diseases because of the pathology itself or its treatment, are at high risk for COVID-19 and its complications. While empirical data are sometimes conflicting, this heightened risk has been confirmed in multiple well-done studies for patients with hematologic malignancies, particularly those with B-cell lymphoid malignancies who received lymphocytotoxic therapies, those with a history of recent hematopoietic stem cell transplant and chimeric antigen receptor T-cell therapy, and, to a lesser degree, those with hemoglobinopathies. Patients with immunosuppression need to have a lower threshold for avoiding indoor public spaces where they are unable to effectively keep a safe distance from others, and wear a high-quality well-fitting mask, especially when community levels are not low. They should receive an enhanced initial vaccine regimen and additional boosting. Therapeutic options are available and immunosuppressed patients are prioritized per the NIH.
Collapse
Affiliation(s)
- Elie A Saade
- Division of Infectious Diseases and HIV Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Case Western Reserve University, Cleveland, OH, USA.
| | - Leila S Hojat
- Division of Infectious Diseases and HIV Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Case Western Reserve University, Cleveland, OH, USA
| | - Praveen Gundelly
- Division of Infectious Diseases and HIV Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Case Western Reserve University, Cleveland, OH, USA
| | - Robert A Salata
- Division of Infectious Diseases and HIV Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
26
|
COVID-19 and venous thromboembolism risk in patients with sickle cell disease. Blood Adv 2022. [DOI: 10.1182/bloodadvances.2022007219
expr 883288724 + 831744770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Abstract
Venous thromboembolism (VTE) is a life-threatening complication observed among patients with sickle cell disease (SCD) and also among those with severe COVID-19 infection. Although prior studies show that patients with SCD are at risk of severe COVID-19 illness, it remains unclear if COVID-19 infection further increases VTE risk for this population. We hypothesized that patients with SCD hospitalized for COVID-19 would have higher VTE rates than those hospitalized for other causes. Using electronic health record data from a multisite research network, TriNetX, we identified 2 groups of patients with SCD hospitalized during 2020: (1) with COVID-19 and (2) without COVID-19. We compared VTE rates using risk ratios estimated based on adjusted Poisson regression model with log link and robust error variances. Of the 281 SCD patients hospitalized with COVID-19 and 4873 SCD patients hospitalized without COVID-19 , 35 (12.46%) and 418 (8.58%) had incident VTE within 6 months of the index hospitalization respectively. After adjusting for differences in baseline characteristics, no significant differences in VTE rates within 6 months were found between the 2 groups (adjusted relative risk, 1.06 [95% confidence interval, 0.79-1.41]). These data suggest that hospitalization with COVID-19 does not further increase VTE risk in patients with SCD.
Collapse
|
27
|
COVID-19 and venous thromboembolism risk in patients with sickle cell disease. Blood Adv 2022; 6. [PMID: 35763429 PMCID: PMC9239700 DOI: 10.1182/bloodadvances.2022007219&set/a 864100454+862542244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Venous thromboembolism (VTE) is a life-threatening complication observed among patients with sickle cell disease (SCD) and also among those with severe COVID-19 infection. Although prior studies show that patients with SCD are at risk of severe COVID-19 illness, it remains unclear if COVID-19 infection further increases VTE risk for this population. We hypothesized that patients with SCD hospitalized for COVID-19 would have higher VTE rates than those hospitalized for other causes. Using electronic health record data from a multisite research network, TriNetX, we identified 2 groups of patients with SCD hospitalized during 2020: (1) with COVID-19 and (2) without COVID-19. We compared VTE rates using risk ratios estimated based on adjusted Poisson regression model with log link and robust error variances. Of the 281 SCD patients hospitalized with COVID-19 and 4873 SCD patients hospitalized without COVID-19 , 35 (12.46%) and 418 (8.58%) had incident VTE within 6 months of the index hospitalization respectively. After adjusting for differences in baseline characteristics, no significant differences in VTE rates within 6 months were found between the 2 groups (adjusted relative risk, 1.06 [95% confidence interval, 0.79-1.41]). These data suggest that hospitalization with COVID-19 does not further increase VTE risk in patients with SCD.
Collapse
|
28
|
Wulftange WJ, Kucukal E, Man Y, An R, Monchamp K, Sevrain CD, Dashora HR, Owusu-Ansah AT, Bode A, Ilich A, Little JA, Key NS, Gurkan UA. Antithrombin-III mitigates thrombin-mediated endothelial cell contraction and sickle red blood cell adhesion in microscale flow. Br J Haematol 2022; 198:893-902. [PMID: 35822297 PMCID: PMC9542057 DOI: 10.1111/bjh.18328] [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: 04/21/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 11/27/2022]
Abstract
Individuals with sickle cell disease (SCD) have persistently elevated thrombin generation that results in a state of systemic hypercoagulability. Antithrombin‐III (ATIII), an endogenous serine protease inhibitor, inhibits several enzymes in the coagulation cascade, including thrombin. Here, we utilize a biomimetic microfluidic device to model the morphology and adhesive properties of endothelial cells (ECs) activated by thrombin and examine the efficacy of ATIII in mitigating the adhesion of SCD patient‐derived red blood cells (RBCs) and EC retraction. Microfluidic devices were fabricated, seeded with ECs, and incubated under physiological shear stress. Cells were then activated with thrombin with or without an ATIII pretreatment. Blood samples from subjects with normal haemoglobin (HbAA) and subjects with homozygous SCD (HbSS) were used to examine RBC adhesion to ECs. Endothelial cell surface adhesion molecule expression and confluency in response to thrombin and ATIII treatments were also evaluated. We found that ATIII pretreatment of ECs reduced HbSS RBC adhesion to thrombin‐activated endothelium. Furthermore, ATIII mitigated cellular contraction and reduced surface expression of von Willebrand factor and vascular cell adhesion molecule‐1 (VCAM‐1) mediated by thrombin. Our findings suggest that, by attenuating thrombin‐mediated EC damage and RBC adhesion to endothelium, ATIII may alleviate the thromboinflammatory manifestations of SCD.
Collapse
Affiliation(s)
- William J Wulftange
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Erdem Kucukal
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yuncheng Man
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ran An
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Karamoja Monchamp
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Charlotte D Sevrain
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Himanshu R Dashora
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Amma T Owusu-Ansah
- Department of Pediatrics, Division of Hematology Oncology, University Hospitals Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| | - Allison Bode
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Anton Ilich
- Division of Hematology and UNC Blood Research Center, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jane A Little
- Division of Hematology and UNC Blood Research Center, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Nigel S Key
- Division of Hematology and UNC Blood Research Center, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Umut A Gurkan
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
29
|
Torres LS, Asada N, Weiss MJ, Trumpp A, Suda T, Scadden DT, Ito K. Recent advances in "sickle and niche" research - Tribute to Dr. Paul S Frenette. Stem Cell Reports 2022; 17:1509-1535. [PMID: 35830837 PMCID: PMC9287685 DOI: 10.1016/j.stemcr.2022.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 10/27/2022] Open
Abstract
In this retrospective, we review the two research topics that formed the basis of the outstanding career of Dr. Paul S. Frenette. In the first part, we focus on sickle cell disease (SCD). The defining feature of SCD is polymerization of the deoxygenated mutant hemoglobin, which leads to a vicious cycle of hemolysis and vaso-occlusion. We survey important discoveries in SCD pathophysiology that have led to recent advances in treatment of SCD. The second part focuses on the hematopoietic stem cell (HSC) niche, the complex microenvironment within the bone marrow that controls HSC function and homeostasis. We detail the cells that constitute this niche, and the factors that these cells use to exert control over hematopoiesis. Here, we trace the scientific paths of Dr. Frenette, highlight key aspects of his research, and identify his most important scientific contributions in both fields.
Collapse
Affiliation(s)
- Lidiane S Torres
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69117 Heidelberg, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Toshio Suda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - David T Scadden
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Einstein Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
30
|
COVID-19 and Venous Thromboembolism Risk in Patients With Sickle Cell Disease. Blood Adv 2022; 6:4408-4412. [PMID: 35763429 PMCID: PMC9239700 DOI: 10.1182/bloodadvances.2022007219] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/17/2022] [Indexed: 12/21/2022] Open
Abstract
Venous thromboembolism (VTE) is a life-threatening complication observed among patients with sickle cell disease (SCD) and also among those with severe COVID-19 infection. Although prior studies show that patients with SCD are at risk of severe COVID-19 illness, it remains unclear if COVID-19 infection further increases VTE risk for this population. We hypothesized that patients with SCD hospitalized for COVID-19 would have higher VTE rates than those hospitalized for other causes. Using electronic health record data from a multisite research network, TriNetX, we identified 2 groups of patients with SCD hospitalized during 2020: (1) with COVID-19 and (2) without COVID-19. We compared VTE rates using risk ratios estimated based on adjusted Poisson regression model with log link and robust error variances. Of the 281 SCD patients hospitalized with COVID-19 and 4873 SCD patients hospitalized without COVID-19 , 35 (12.46%) and 418 (8.58%) had incident VTE within 6 months of the index hospitalization respectively. After adjusting for differences in baseline characteristics, no significant differences in VTE rates within 6 months were found between the 2 groups (adjusted relative risk, 1.06 [95% confidence interval, 0.79-1.41]). These data suggest that hospitalization with COVID-19 does not further increase VTE risk in patients with SCD.
Collapse
|
31
|
Ionescu F, Anusim N, Zimmer M, Jaiyesimi I. Venous thromboembolism prophylaxis in hospitalized sickle cell disease and sickle cell trait patients. Eur J Haematol Suppl 2022; 109:282-288. [PMID: 35617049 DOI: 10.1111/ejh.13807] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Sickle trait (Hb SA) or sickle disease (Hb SS) carries increased risk of venous thromboembolism (VTE). Hb SS patients are young and lack common comorbid conditions that qualify them for VTE prophylaxis (VTEP). METHODS Retrospective, multicenter analysis of Hb SS/Hb SA adult patients between January 2013 and December 2018. RESULTS There were 803 Hb SA (525 patients) and 1020 Hb SS admissions (262 patients). VTEP use was similar between Hb SA and controls (42% vs. 46%; p-value = .06) and Hb SS and controls (45% vs. 42%; p-value = .13). Hb SS/Hb SA patients more frequently received more than half of prescribed doses of VTEP. In multivariate analysis, increasing age and longer hospitalizations were positive predictors. Odds of VTEP use varied with treatment site for Hb SS patients, whereas comorbid conditions, admission hemoglobin and platelet count were not predictive. By contrast, in Hb SA patients, comorbid conditions, higher admission hemoglobin, and higher admission platelet counts raised the odds of VTEP being offered. CONCLUSIONS VTEP is underused in Hb SS/Hb SA patients. There may be a trend toward offering more VTEP in Hb SS disease, but not in Hb SA patients, where VTEP prescribing is driven by comorbid conditions rather than genotype. Patient compliance does not appear to play a major role, but intercenter variability suggests provider education may improve VTEP use.
Collapse
Affiliation(s)
- Filip Ionescu
- Department of Internal Medicine, Beaumont Health System, OUWB School of Medicine, Royal Oak, Michigan, USA.,H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, Florida, USA
| | - Nwabundo Anusim
- Department of Hematology and Oncology, Dell Medical School, University of Texas in Austin, Austin, Texas, USA.,Department of Hematology-Oncology, Beaumont Health System, OUWB School of Medicine, Royal Oak, Michigan, USA
| | - Markie Zimmer
- Department of Internal Medicine, Beaumont Health System, OUWB School of Medicine, Royal Oak, Michigan, USA
| | - Ishmael Jaiyesimi
- Department of Hematology-Oncology, Beaumont Health System, OUWB School of Medicine, Royal Oak, Michigan, USA
| |
Collapse
|
32
|
Notariale R, Perrone P, Mele L, Lettieri G, Piscopo M, Manna C. Olive Oil Phenols Prevent Mercury-Induced Phosphatidylserine Exposure and Morphological Changes in Human Erythrocytes Regardless of Their Different Scavenging Activity. Int J Mol Sci 2022; 23:ijms23105693. [PMID: 35628502 PMCID: PMC9147954 DOI: 10.3390/ijms23105693] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 02/05/2023] Open
Abstract
Phosphatidylserine (PS) translocation to the external membrane leaflet represents a key mechanism in the pathophysiology of human erythrocytes (RBC) acting as an "eat me" signal for the removal of aged/stressed cells. Loss of physiological membrane asymmetry, however, can lead to adverse effects on the cardiovascular system, activating a prothrombotic activity. The data presented indicate that structurally related olive oil phenols prevent cell alterations induced in intact human RBC exposed to HgCl2 (5-40 µM) or Ca2+ ionophore (5 µM), as measured by hallmarks including PS exposure, reactive oxygen species generation, glutathione depletion and microvesicles formation. The protective effect is observed in a concentration range of 1-30 µM, hydroxytyrosol being the most effective; its in vivo metabolite homovanillic alcohol still retains the biological activity of its dietary precursor. Significant protection is also exerted by tyrosol, in spite of its weak scavenging activity, indicating that additional mechanisms are involved in the protective effect. When RBC alterations are mediated by an increase in intracellular calcium, the protective effect is observed at higher concentrations, indicating that the selected phenols mainly act on Ca2+-independent mechanisms, identified as protection of glutathione depletion. Our findings strengthen the nutritional relevance of olive oil bioactive compounds in the claimed health-promoting effects of the Mediterranean Diet.
Collapse
Affiliation(s)
- Rosaria Notariale
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.N.); (P.P.)
| | - Pasquale Perrone
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.N.); (P.P.)
| | - Luigi Mele
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Gennaro Lettieri
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.L.); (M.P.)
| | - Marina Piscopo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.L.); (M.P.)
| | - Caterina Manna
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.N.); (P.P.)
- Correspondence:
| |
Collapse
|
33
|
Feugray G, Kasonga F, Grall M, Benhamou Y, Bobée-Schneider V, Buchonnet G, Daliphard S, Le Cam Duchez V, Lahary A, Billoir P. Assessment of Reticulocyte and Erythrocyte Parameters From Automated Blood Counts in Vaso-Occlusive Crisis on Sickle Cell Disease. Front Med (Lausanne) 2022; 9:858911. [PMID: 35492334 PMCID: PMC9044919 DOI: 10.3389/fmed.2022.858911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/21/2022] [Indexed: 12/18/2022] Open
Abstract
Sickle cell disease is a complex genetic disease involving cell adhesion between red blood cells, white blood cells, platelets and endothelial cells, inducing painful vaso-occlusive crisis (VOC). We assessed reticulocyte and erythrocyte parameters in a cohort of confirmed SCD patients, and investigated whether a combination of these routine laboratory biomarkers of haemolysis could be used to predict VOC development. Reticulocyte and erythrocyte parameters were evaluated using the Sysmex XN-9000 analyser. A total of 98 patients with SCD were included, 72 in steady state and 26 in VOC. Among the 72 patients in steady state, 22 developed a VOC in the following year (median: 3 months [2-6]). The following parameters were increased in SCD patients with VOC development compared to SCD patients without VOC development in the following year: reticulocyte count (94.6 109/L [67.8-128] vs. 48.4 109/L [24.9-87.5]), immature reticulocyte count (259 109/L [181-334] vs. 152 109/L [129-208]) reticulocyte/immature reticulocyte fraction (IRF) ratio (6.63 109/(L*%) [4.67-9.56] vs. 4.94 109/(L*%) [3.96-6.61]), and medium fluorescence reticulocytes (MFR) (19.9% [17.4-20.7] vs. 17.1% [15.95-19.75]). The association of a reticulocyte count of >189.4 109/L and an MFR of >19.75% showed a sensitivity of 81.8% and a specificity of 88% to predict VOC development in the following year. Based on our findings, a combination of routine laboratory biomarkers, as reticulocyte count, immature reticulocyte count and fluorescent reticulocyte fraction at steady state, could be used to predict VOC development in SCD.
Collapse
Affiliation(s)
- Guillaume Feugray
- General Biochemistry, Normandie Univ, Rouen University, INSERM U1096, CHU Rouen, Rouen, France
| | | | | | - Ygal Benhamou
- Department of Internal Medecine, Normandie Univ, Rouen University, INSERM U1096, CHU Rouen, Rouen, France
| | | | | | | | - Véronique Le Cam Duchez
- Normandie Univ, Rouen University, INSERM U1096, CHU Rouen, Vascular Hemostasis Unit, Rouen, France
| | | | - Paul Billoir
- Normandie Univ, Rouen University, INSERM U1096, CHU Rouen, Vascular Hemostasis Unit, Rouen, France
| |
Collapse
|
34
|
Tentolouris A, Stafylidis C, Siafarikas C, Dimopoulou M, Makrodimitri S, Bousi S, Papalexis P, Damaskos C, Trakas N, Sklapani P, Spandidos D, Georgakopoulou V. Favorable outcomes of patients with sickle cell disease hospitalized due to COVID‑19: A report of three cases. Exp Ther Med 2022; 23:338. [PMID: 35401804 PMCID: PMC8988160 DOI: 10.3892/etm.2022.11268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/11/2022] [Indexed: 11/06/2022] Open
Abstract
Sickle cell disease (SCD) is one of the most frequent and severe monogenic disorders, affecting millions of individuals worldwide. SCD represents a fatal hematological illness, characterized by veno-occlusive events and hemolytic anemia. Hemolytic anemia is caused by abnormal sickle-shaped erythrocytes, which induce parenchymal destruction and persistent organ damage, resulting in considerable morbidity and mortality. During the coronavirus disease 2019 (COVID-19) pandemic, patients with SCD were characterized as a ‘high-risk’ group due to their compromised immune system, caused by functional hyposplenism, as well as systemic vasculopathy. COVID-19 is characterized by endothelial damage and a procoagulant condition. The present study describes the clinical features, management and outcomes of 3 patients with SCD who were hospitalized due to COVID-19, who all had favorable outcomes despite the complications.
Collapse
Affiliation(s)
- Anastasios Tentolouris
- First Department of Propedeutic and Internal Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Stafylidis
- First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Siafarikas
- First Department of Propedeutic and Internal Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Dimopoulou
- Hemοglobinopathies Reference Center, Laiko General Hospital, 11527 Athens, Greece
| | - Sotiria Makrodimitri
- Department of Infectious Diseases‑COVID‑19 Unit, Laiko General Hospital, 11527 Athens, Greece
| | - Stelios Bousi
- Renal Transplantation Unit, Laiko General Hospital, 11527 Athens, Greece
| | - Petros Papalexis
- Unit of Endocrinology, First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Damaskos
- Renal Transplantation Unit, Laiko General Hospital, 11527 Athens, Greece
| | - Nikolaos Trakas
- Department of Biochemistry, Sismanogleio Hospital, 15126 Athens, Greece
| | - Pagona Sklapani
- Department of Cytology, Mitera Hospital, 15123 Athens, Greece
| | - Demetrios Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | | |
Collapse
|
35
|
Forté S, De Luna G, Abdulrehman J, Fadiga N, Pestrin O, Pham Hung d’Alexandry d’Orengiani AL, Aneke JC, Guillet H, Budhram D, Habibi A, Ward R, Bartolucci P, Kuo KHM. Thromboprophylaxis Reduced Venous Thromboembolism in Sickle Cell Patients with Central Venous Access Devices: A Retrospective Cohort Study. J Clin Med 2022; 11:jcm11051193. [PMID: 35268283 PMCID: PMC8910838 DOI: 10.3390/jcm11051193] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 12/16/2022] Open
Abstract
Sickle cell disease (SCD) induces a chronic prothrombotic state. Central venous access devices (CVADs) are commonly used for chronic transfusions and iron chelation in this population. CVADs are an additional venous thromboembolism (VTE) risk factor. The role of thromboprophylaxis in this setting is uncertain. The objectives are: (1) to determine whether thromboprophylaxis reduces VTE risk in SCD patients with CVAD and (2) to explore characteristics associated with VTE risk. We identified adults with SCD and CVAD intended for chronic use (≥3 months) at two comprehensive SCD centers. Thromboprophylaxis presence; type; intensity; and patient-, catheter-, and treatment-related VTE risk factors were recorded. Among 949 patients, 49 had a CVAD (25 without and 24 with VTE prophylaxis). Thromboprophylaxis type and intensity varied widely. Patients without thromboprophylaxis had higher VTE rates (rate ratio (RR) = 4.0 (95% confidence interval: 1.2−12.6), p = 0.02). Hydroxyurea was associated with lower VTE rates (RR = 20.5 (6.4−65.3), p < 0.001). PICC lines and Vortex and Xcela Power implantable devices were associated with higher rates compared with Port-a-Cath (RR = 5.8 (1.3−25.9), p = 0.02, and RR = 58.2 (15.0−225.0), p < 0.001, respectively). Thromboprophylaxis, hydroxyurea, and CVAD subtype were independently associated with VTE. The potentially protective role of thromboprophylaxis and hydroxyurea for VTE prevention in patients with SCD and CVAD merits further exploration.
Collapse
Affiliation(s)
- Stéphanie Forté
- Division of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C4, Canada; (S.F.); (J.A.); (N.F.); (O.P.); (J.C.A.); (D.B.); (R.W.)
| | - Gonzalo De Luna
- Sickle Cell Referral Center, Department of Internal Medicine, Henri Mondor University Hospital, UPEC, APHP, 94000 Créteil, France; (G.D.L.); (A.-L.P.H.d.d.); (H.G.); (A.H.); (P.B.)
| | - Jameel Abdulrehman
- Division of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C4, Canada; (S.F.); (J.A.); (N.F.); (O.P.); (J.C.A.); (D.B.); (R.W.)
| | - Nafanta Fadiga
- Division of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C4, Canada; (S.F.); (J.A.); (N.F.); (O.P.); (J.C.A.); (D.B.); (R.W.)
| | - Olivia Pestrin
- Division of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C4, Canada; (S.F.); (J.A.); (N.F.); (O.P.); (J.C.A.); (D.B.); (R.W.)
| | - Anne-Laure Pham Hung d’Alexandry d’Orengiani
- Sickle Cell Referral Center, Department of Internal Medicine, Henri Mondor University Hospital, UPEC, APHP, 94000 Créteil, France; (G.D.L.); (A.-L.P.H.d.d.); (H.G.); (A.H.); (P.B.)
| | - John Chinawaeze Aneke
- Division of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C4, Canada; (S.F.); (J.A.); (N.F.); (O.P.); (J.C.A.); (D.B.); (R.W.)
| | - Henri Guillet
- Sickle Cell Referral Center, Department of Internal Medicine, Henri Mondor University Hospital, UPEC, APHP, 94000 Créteil, France; (G.D.L.); (A.-L.P.H.d.d.); (H.G.); (A.H.); (P.B.)
| | - Dalton Budhram
- Division of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C4, Canada; (S.F.); (J.A.); (N.F.); (O.P.); (J.C.A.); (D.B.); (R.W.)
| | - Anoosha Habibi
- Sickle Cell Referral Center, Department of Internal Medicine, Henri Mondor University Hospital, UPEC, APHP, 94000 Créteil, France; (G.D.L.); (A.-L.P.H.d.d.); (H.G.); (A.H.); (P.B.)
| | - Richard Ward
- Division of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C4, Canada; (S.F.); (J.A.); (N.F.); (O.P.); (J.C.A.); (D.B.); (R.W.)
| | - Pablo Bartolucci
- Sickle Cell Referral Center, Department of Internal Medicine, Henri Mondor University Hospital, UPEC, APHP, 94000 Créteil, France; (G.D.L.); (A.-L.P.H.d.d.); (H.G.); (A.H.); (P.B.)
- Laboratoire D’Excellence, GRex, Institut Mondor, INSERM U955 Equipe 2, 94000 Créteil, France
| | - Kevin H. M. Kuo
- Division of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C4, Canada; (S.F.); (J.A.); (N.F.); (O.P.); (J.C.A.); (D.B.); (R.W.)
- Correspondence:
| |
Collapse
|
36
|
Venous thromboembolism in pediatric patients with sickle cell disease: A north American survey on experience and management approaches of pediatric hematologists. Thromb Res 2022; 211:133-139. [DOI: 10.1016/j.thromres.2022.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/12/2022] [Accepted: 01/31/2022] [Indexed: 11/19/2022]
|
37
|
Rankine-Mullings AE. Ulcerative colitis in patients with sickle cell disease: a rare but important co-morbidity. Paediatr Int Child Health 2022; 42:1-4. [PMID: 35694868 DOI: 10.1080/20469047.2022.2084964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
COVID-19: Coronavirus disease 2019; HIC: high-income countries; IBD: inflammatory bowel disease; LMIC: low- and middle-income countries; PUCAL: paediatric ulcerative colitis activity index; SCD: sickle cell disease; UC: ulcerative colitis.
Collapse
Affiliation(s)
- Angela E Rankine-Mullings
- Sickle Cell Unit, Caribbean Institute for Health Research, The University of the West Indies, Kingston, Jamaica
| |
Collapse
|
38
|
Kalff H, Cario H, Holzhauer S. Iron deficiency anemia and thrombosis risk in children-revisiting an old hypothesis. Front Pediatr 2022; 10:926925. [PMID: 35979407 PMCID: PMC9376258 DOI: 10.3389/fped.2022.926925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Iron deficiency anemia has a high prevalence in children and has repeatedly been implicated as a risk factor for arterial and venous thrombosis. As an effective therapy for iron deficiency anemia is available, understanding the association between this form of anemia and the potentially severe thrombosis phenotype is of major clinical interest. Recent findings shed light on pathophysiology of hypercoagulability resulting from iron-restricted erythropoiesis. Specifically, an animal model of induced iron deficiency allowed identifying multiple mechanisms, by which iron deficiency anemia results in increased thrombus formation and thrombus progression both in arterial and venous thrombosis. These findings complement and support conclusions derived from clinical data. The purpose of this mini review is to summarize current evidence on the association of iron deficiency anemia and thrombosis. We want to increase the awareness of iron deficiency as a risk factor for thrombosis in the pediatric population. We discuss how novel pathophysiological concepts can be translated into the clinical settings and suggest clinical studies on prevention and treatment strategies in high-risk patient groups.
Collapse
Affiliation(s)
- Hannah Kalff
- Department of Pediatric Hematology and Oncology, Charité University Medicine, Berlin, Germany
| | - Holger Cario
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Susanne Holzhauer
- Department of Pediatric Hematology and Oncology, Charité University Medicine, Berlin, Germany
| |
Collapse
|
39
|
Nader E, Garnier Y, Connes P, Romana M. Extracellular Vesicles in Sickle Cell Disease: Plasma Concentration, Blood Cell Types Origin Distribution and Biological Properties. Front Med (Lausanne) 2021; 8:728693. [PMID: 34490315 PMCID: PMC8417591 DOI: 10.3389/fmed.2021.728693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/30/2021] [Indexed: 01/08/2023] Open
Abstract
Prototype of monogenic disorder, sickle cell disease (SCD) is caused by a unique single mutation in the β-globin gene, leading to the production of the abnormal hemoglobin S (HbS). HbS polymerization in deoxygenated condition induces the sickling of red blood cells (RBCs), which become less deformable and more fragile, and thus prone to lysis. In addition to anemia, SCD patients may exhibit a plethora of clinical manifestations ranging from acute complications such as the frequent and debilitating painful vaso-occlusive crisis to chronic end organ damages. Several interrelated pathophysiological processes have been described, including impaired blood rheology, increased blood cell adhesion, coagulation, inflammation and enhanced oxidative stress among others. During the last two decades, it has been shown that extracellular vesicles (EVs), defined as cell-derived anucleated particles delimited by a lipid bilayer, and comprising small EVs (sEVs) and medium/large EVs (m/lEVs); are not only biomarkers but also subcellular actors in SCD pathophysiology. Plasma concentration of m/lEVs, originated mainly from RBCs and platelets (PLTs) but also from the other blood cell types, is higher in SCD patients than in healthy controls. The concentration and the density of externalized phosphatidylserine of those released from RBCs may vary according to clinical status (crisis vs. steady state) and treatment (hydroxyurea). Besides their procoagulant properties initially described, RBC-m/lEVs may promote inflammation through their effects on monocytes/macrophages and endothelial cells. Although less intensely studied, sEVs plasma concentration is increased in SCD and these EVs may cause endothelial damages. In addition, sEVs released from activated PLTs trigger PLT-neutrophil aggregation involved in lung vaso-occlusion in sickle mice. Altogether, these data clearly indicate that EVs are both biomarkers and bio-effectors in SCD, which deserve further studies.
Collapse
Affiliation(s)
- Elie Nader
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Team "Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France
| | - Yohann Garnier
- Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| | - Philippe Connes
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Team "Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France
| | - Marc Romana
- Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| |
Collapse
|
40
|
Sharathkumar AA, Faustino EVS, Takemoto CM. How we approach thrombosis risk in children with COVID-19 infection and MIS-C. Pediatr Blood Cancer 2021; 68:e29049. [PMID: 33955167 PMCID: PMC8206673 DOI: 10.1002/pbc.29049] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/04/2021] [Accepted: 03/22/2021] [Indexed: 01/08/2023]
Abstract
Thrombosis within the microvasculature and medium to large vessels is a serious and common complication among critically ill individuals with coronavirus disease 2019 (COVID-19). While children are markedly less likely to develop severe disease than adults, they remain at risk for thrombosis during acute infection and with the post-acute inflammatory illness termed multisystem inflammatory syndrome in children. Significant knowledge deficits in understanding COVID-19-associated coagulopathy and thrombotic risk pose clinical challenges for pediatric providers who must incorporate expert opinion and personal experience to manage individual patients. We discuss clinical scenarios to provide framework for characterizing thrombosis risk and thromboprophylaxis in children with COVID-19.
Collapse
Affiliation(s)
- Anjali A. Sharathkumar
- Stead Family Department of Pediatrics, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - E. Vincent S. Faustino
- Section of Pediatric Critical Care Medicine, Department of PediatricsYale School of MedicineNew HavenConnecticutUSA
| | - Clifford M. Takemoto
- Division of Clinical HematologySt. Jude Children's Research HospitalMemphisTennesseeUSA
| |
Collapse
|
41
|
Conran N, Embury SH. Sickle cell vaso-occlusion: The dialectic between red cells and white cells. Exp Biol Med (Maywood) 2021; 246:1458-1472. [PMID: 33794696 DOI: 10.1177/15353702211005392] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The pathophysiology of sickle cell anemia, a hereditary hemoglobinopathy, has fascinated clinicians and scientists alike since its description over 100 years ago. A single gene mutation in the HBB gene results in the production of abnormal hemoglobin (Hb) S, whose polymerization when deoxygenated alters the physiochemical properties of red blood cells, in turn triggering pan-cellular activation and pathological mechanisms that include hemolysis, vaso-occlusion, and ischemia-reperfusion to result in the varied and severe complications of the disease. Now widely regarded as an inflammatory disease, in recent years attention has included the role of leukocytes in vaso-occlusive processes in view of the part that these cells play in innate immune processes, their inherent ability to adhere to the endothelium when activated, and their sheer physical and potentially obstructive size. Here, we consider the role of sickle red blood cell populations in elucidating the importance of adhesion vis-a-vis polymerization in vaso-occlusion, review the direct adhesion of sickle red cells to the endothelium in vaso-occlusive processes, and discuss how red cell- and leukocyte-centered mechanisms are not mutually exclusive. Given the initial clinical success of crizanlizumab, a specific anti-P selectin therapy, we suggest that it is appropriate to take a holistic approach to understanding and exploring the complexity of vaso-occlusive mechanisms and the adhesive roles of the varied cell types, including endothelial cells, platelets, leukocytes, and red blood cells.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology Center, University of Campinas-UNICAMP, Barão Geraldo 13083-8, Campinas, SP, Brazil
| | | |
Collapse
|
42
|
Karki NR, Kutlar A. P-Selectin Blockade in the Treatment of Painful Vaso-Occlusive Crises in Sickle Cell Disease: A Spotlight on Crizanlizumab. J Pain Res 2021; 14:849-856. [PMID: 33833562 PMCID: PMC8019662 DOI: 10.2147/jpr.s278285] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/13/2021] [Indexed: 01/01/2023] Open
Abstract
Microvascular vaso-occlusion driven pain crisis is the hallmark of sickle cell disease with profound morbidity and increased mortality. Selectins, most notably P-selectins have an integral role in this phenomenon. P-selection was first identified in 1989. In 2019, after 3 decades of basic, translational, and clinical work with this pathway, the US Food and Drug Administration approved a P-selectin antibody, crizanlizumab to reduce frequency of pain crisis in patients more than 16 years with sickle cell disease. We review the fundamentals of P-selectin pathobiology, P-selectin blocking agents, clinical data with the use of crizanlizumab and prospects of this novel class of drugs in the context of other treatments for painful vaso-occlusive episodes.
Collapse
Affiliation(s)
- Nabin Raj Karki
- Division of Hematology/Oncology, Augusta University, Augusta, GA, USA
| | - Abdullah Kutlar
- Division of Hematology/Oncology, Augusta University, Augusta, GA, USA
| |
Collapse
|
43
|
Hopp MT, Imhof D. Linking Labile Heme with Thrombosis. J Clin Med 2021; 10:427. [PMID: 33499296 PMCID: PMC7865584 DOI: 10.3390/jcm10030427] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Thrombosis is one of the leading causes of death worldwide. As such, it also occurs as one of the major complications in hemolytic diseases, like hemolytic uremic syndrome, hemorrhage and sickle cell disease. Under these conditions, red blood cell lysis finally leads to the release of large amounts of labile heme into the vascular compartment. This, in turn, can trigger oxidative stress and proinflammatory reactions. Moreover, the heme-induced activation of the blood coagulation system was suggested as a mechanism for the initiation of thrombotic events under hemolytic conditions. Studies of heme infusion and subsequent thrombotic reactions support this assumption. Furthermore, several direct effects of heme on different cellular and protein components of the blood coagulation system were reported. However, these effects are controversially discussed or not yet fully understood. This review summarizes the existing reports on heme and its interference in coagulation processes, emphasizing the relevance of considering heme in the context of the treatment of thrombosis in patients with hemolytic disorders.
Collapse
Affiliation(s)
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany;
| |
Collapse
|