51
|
Pulmonary Hypertension in Children with Sickle Cell Disease: a Review of the Current Literature. CURRENT PEDIATRICS REPORTS 2019. [DOI: 10.1007/s40124-019-00188-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
52
|
Houwing ME, de Pagter PJ, van Beers EJ, Biemond BJ, Rettenbacher E, Rijneveld AW, Schols EM, Philipsen JNJ, Tamminga RYJ, van Draat KF, Nur E, Cnossen MH. Sickle cell disease: Clinical presentation and management of a global health challenge. Blood Rev 2019; 37:100580. [PMID: 31128863 DOI: 10.1016/j.blre.2019.05.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/17/2019] [Accepted: 05/17/2019] [Indexed: 01/12/2023]
Abstract
Sickle cell disease is an autosomal recessive, multisystem disorder, characterised by chronic haemolytic anaemia, painful episodes of vaso-occlusion, progressive organ failure and a reduced life expectancy. Sickle cell disease is the most common monogenetic disease, with millions affected worldwide. In well-resourced countries, comprehensive care programs have increased life expectancy of sickle cell disease patients, with almost all infants surviving into adulthood. Therapeutic options for sickle cell disease patients are however, still scarce. Predictors of sickle cell disease severity and a better understanding of pathophysiology and (epi)genetic modifiers are warranted and could lead to more precise management and treatment. This review provides an extensive summary of the pathophysiology and management of sickle cell disease and encompasses the characteristics, complications and current and future treatment options of the disease.
Collapse
Affiliation(s)
- M E Houwing
- Department of Paediatric Haematology, Erasmus University Medical Center - Sophia Children's Hospital, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| | - P J de Pagter
- Department of Paediatric Haematology, Erasmus University Medical Center - Sophia Children's Hospital, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| | - E J van Beers
- Department of Internal Medicine and Dermatology, Van Creveldkliniek, University Medical Center Utrecht, Internal mail no C.01.412, 3508, GA, Utrecht, the Netherlands.
| | - B J Biemond
- Department of Internal Medicine and Clinical Haematology, Amsterdam University Medical Centers, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands.
| | - E Rettenbacher
- Department of Paediatric Haematology, Radboud University Medical Center - Amalia Children's Hospital, Geert Grooteplein Zuid 10, 6500, HB, Nijmegen, the Netherlands.
| | - A W Rijneveld
- Department of Haematology, Erasmus University Medical Center, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| | - E M Schols
- Department of Haematology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, the Netherlands.
| | - J N J Philipsen
- Department of Cell Biology, Erasmus University Medical Center, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| | - R Y J Tamminga
- Department of Paediatric Oncology and Haematology, University Medical Center Groningen - Beatrix Children's Hospital, Postbus 30001, 9700, RB, Groningen, the Netherlands..
| | - K Fijn van Draat
- Department of Paediatric Haematology, Amsterdam University Medical Centers - Emma Children's Hospital, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Department of Plasma Proteins, Sanquin Research, the Netherlands.
| | - E Nur
- Department of Internal Medicine and Clinical Haematology, Amsterdam University Medical Centers, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands.
| | - M H Cnossen
- Department of Paediatric Haematology, Erasmus University Medical Center - Sophia Children's Hospital, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| |
Collapse
|
53
|
Hammoudi N, Lionnet F, Redheuil A, Montalescot G. Cardiovascular manifestations of sickle cell disease. Eur Heart J 2019; 41:1365-1373. [DOI: 10.1093/eurheartj/ehz217] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 03/04/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023] Open
Abstract
Abstract
Sickle cell disease (SCD) is the most frequent genetic haemoglobinopathy worldwide. Early childhood mortality has dramatically decreased in high-income countries, and most patients now survive beyond the 5th decade. However, in the aging SCD population, the morbidity related to chronic organ damage, especially kidney and heart, has become a major concern. While pulmonary hypertension has attracted most attention, it appears that this condition is frequently linked to left heart failure (HF). Accordingly, SCD-associated cardiomyopathy is emerging as a major cause of reduced quality of life and early mortality in these patients. The diagnosis of this particular phenotype of high-output HF is challenging. Exercise intolerance and dyspnoea in SCD patients are linked to multiple causes including chronic anaemia. Moreover, echocardiographic features are unusual and can be misinterpreted. The classical diagnosis algorithm for HF is generally not suitable in SCD patients, and HF is poorly recognized and mostly diagnosed at a late congestive stage in routine practice. Such patients need to be identified at an earlier stage of myocardial dysfunction via improved phenotyping. This constitutes the first step towards further investigations in SCD needed to improve the prognosis and the quality of life. This article provides an updated review of the recent advances in the pathophysiology and diagnosis, and in addition, perspectives of new therapeutic approaches in SCD-related cardiac manifestations.
Collapse
Affiliation(s)
- Nadjib Hammoudi
- Sorbonne Université, Inserm, Institute of Cardiometabolism and Nutrition (ICAN), ACTION Study Group and Department of Cardiology, Institute of Cardiology (AP-HP), Hôpital Pitié- Salpêtrière, Boulevard de l'hôpital, Paris F-75013, France
| | - François Lionnet
- Sorbonne Université, Department of Internal Medicine, centre de référence de la drépanocytose (AP-HP), Centre Hospitalier Universitaire Tenon, rue de la Chine, 75020 Paris, France
| | - Alban Redheuil
- Department of Cardiovascular Imaging, Interventional and Thoracic Radiology (DICVRIT), Hôpital Pitié- Salpêtrière, Boulevard de l'hôpital, Sorbonne Université, Inserm, CNRS 7371, Laboratoire d'Imagerie Biomédicale, Institute of Cardiometabolism and Nutrition (ICAN), Institute of Cardiology (AP-HP), Paris F-75013, France
| | - Gilles Montalescot
- Sorbonne Université, Inserm, Institute of Cardiometabolism and Nutrition (ICAN), ACTION Study Group and Department of Cardiology, Institute of Cardiology (AP-HP), Hôpital Pitié- Salpêtrière, Boulevard de l'hôpital, Paris F-75013, France
| |
Collapse
|
54
|
Successful Use of Pulmonary Vasodilators in Acute Chest Syndrome Complicated by Persistent Right Ventricular Failure. Case Rep Cardiol 2019; 2019:4681392. [PMID: 31149361 PMCID: PMC6501236 DOI: 10.1155/2019/4681392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/25/2019] [Accepted: 04/01/2019] [Indexed: 02/02/2023] Open
Abstract
Pulmonary hypertension (PH) is a known consequence of sickle cell disease (SCD) and is associated with increased mortality and more frequent episodes of acute chest syndrome (ACS). Pulmonary pressures are known to increase during ACS, and right ventricular (RV) failure has been described as a significant cause of mortality in this condition. Management of ACS includes exchange transfusion, pain control, and prevention of hypovolemia and hypoxemia. However, in patients with a history of precapillary PH in whom ACS is complicated by persistent RV failure and cardiogenic shock, RV afterload reduction with pulmonary vasodilators may be an effective treatment strategy. Here, we present a case of a young man with SCD-associated PH and ACS who was successfully managed with inhaled and oral pulmonary vasodilators in the setting of persistent elevations in pulmonary vascular resistance leading to acute RV failure and cardiogenic shock.
Collapse
|
55
|
Carstens GR, Paulino BBA, Katayama EH, Amato-Lourenço LF, Fonseca GH, Souza R, Aiello VD, Mauad T. Clinical relevance of pulmonary vasculature involvement in sickle cell disease. Br J Haematol 2019; 185:317-326. [PMID: 30739309 DOI: 10.1111/bjh.15795] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/14/2018] [Indexed: 12/21/2022]
Abstract
Pulmonary complications are frequent in patients with sickle cell disease (SCD), but few studies have described lung pathology in SCD. We studied the lung tissue of 30 deceased SCD patients (1994-2012). Demographics, genotype, clinical characteristics, cause of death and associated conditions are presented. We quantified the presence of pulmonary arterial changes, thrombosis and venous thickening. Alveolar capillary abnormalities were demonstrated using CD34 expression and confocal microscopy. Autopsy and echocardiography reports were reviewed to classify heart abnormalities. Tissue expression of markers of endothelial activation (vascular cell adhesion molecule 1, intercellular adhesion molecule 1 and vascular endothelial growth factor) was quantified in pulmonary vessels. Median age was 33 years; genotype was SS in 19, SC in 7 and Sβ in 4, and there were 18 males. Hypertensive arterial changes were present in 76% of the patients, recent thrombosis in 80% and old thrombosis in 43%. Venous thickening was present in 23% and pulmonary capillary haemangiomatosis foci in 87%. Ten percent of the patients presented right ventricular hypertrophy. There was no increased expression of endothelial activation markers when compared to controls. SCD affects the whole pulmonary vascular tree and reflects the multiple burden on lung vasculature imposed by the disease upon time.
Collapse
Affiliation(s)
- German R Carstens
- Department of Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
- Hospital Eduardo Schütz Schroeder, Puerto Montt, Chile
| | - Bianca B A Paulino
- Department of Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Edgard H Katayama
- Department of Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Luis F Amato-Lourenço
- Department of Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Guilherme H Fonseca
- Department of Haematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Rogerio Souza
- Pulmonary Department, Heart Institute (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Vera D Aiello
- Laboratory of Pathology, Heart Institute (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Thais Mauad
- Department of Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
56
|
Fort R. Recommendations for the use of red blood cell exchange in sickle cell disease. Transfus Apher Sci 2019; 58:128-131. [PMID: 30879904 DOI: 10.1016/j.transci.2019.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Sickle cell disease (SCD) is a genetic disorder characterised by a single mutation of the beta globin gene, causing the production of an abnormal haemoglobin called sickle haemoglobin (HbS). In its deoxygenated form, HbS polymerises, causing major rheological disorders, which presents clinically as periodic vaso-occlusive crises, chronic haemolysis and chronic vascular dysfunction. Patients often resort to a background treatment, and transfusion remains the cornerstone in the management of the disease, significantly reducing morbidity and mortality. The aim of red blood cell exchange (RBCX) is to improve tissue oxygenation by increasing haemoglobin levels while lowering HbS levels. RBCX can be performed by manual or automated exchange, and each technique has its own set of advantages and disadvantages. This article will outline the transfusion indications for the main complications of SCD, as well as the most appropriate strategy to use.
Collapse
Affiliation(s)
- Romain Fort
- Department of Internal Medicine, CHU Edouard Herriot, Lyon, France; Laboratoire LIBM EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard, Lyon, France; Laboratoire d'Excellence du Globule Rouge (LABEX GR-Ex), PRES Sorbonne, Paris, France.
| |
Collapse
|
57
|
Simonneau G, Montani D, Celermajer DS, Denton CP, Gatzoulis MA, Krowka M, Williams PG, Souza R. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur Respir J 2019; 53:13993003.01913-2018. [PMID: 30545968 PMCID: PMC6351336 DOI: 10.1183/13993003.01913-2018] [Citation(s) in RCA: 2545] [Impact Index Per Article: 424.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 10/09/2018] [Indexed: 12/13/2022]
Abstract
Since the 1st World Symposium on Pulmonary Hypertension (WSPH) in 1973, pulmonary hypertension (PH) has been arbitrarily defined as mean pulmonary arterial pressure (mPAP) ≥25 mmHg at rest, measured by right heart catheterisation. Recent data from normal subjects has shown that normal mPAP was 14.0±3.3 mmHg. Two standard deviations above this mean value would suggest mPAP >20 mmHg as above the upper limit of normal (above the 97.5th percentile). This definition is no longer arbitrary, but based on a scientific approach. However, this abnormal elevation of mPAP is not sufficient to define pulmonary vascular disease as it can be due to an increase in cardiac output or pulmonary arterial wedge pressure. Thus, this 6th WSPH Task Force proposes to include pulmonary vascular resistance ≥3 Wood Units in the definition of all forms of pre-capillary PH associated with mPAP >20 mmHg. Prospective trials are required to determine whether this PH population might benefit from specific management. Regarding clinical classification, the main Task Force changes were the inclusion in group 1 of a subgroup “pulmonary arterial hypertension (PAH) long-term responders to calcium channel blockers”, due to the specific prognostic and management of these patients, and a subgroup “PAH with overt features of venous/capillaries (pulmonary veno-occlusive disease/pulmonary capillary haemangiomatosis) involvement”, due to evidence suggesting a continuum between arterial, capillary and vein involvement in PAH. State of the art and research perspectives of haemodynamic definitions and clinical classification of pulmonary hypertensionhttp://ow.ly/TJeR30mgWKj
Collapse
Affiliation(s)
- Gérald Simonneau
- Université Paris-Sud, AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S999, LabEx LERMIT, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - David Montani
- Université Paris-Sud, AP-HP, Centre de Référence de l'Hypertension Pulmonaire, Service de Pneumologie, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S999, LabEx LERMIT, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - David S Celermajer
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Christopher P Denton
- Centre for Rheumatology, Royal Free Campus, University College London, London, UK
| | - Michael A Gatzoulis
- Adult Congenital Heart Centre and National Centre for Pulmonary Hypertension, Royal Brompton and Harefield NHS Trust, and the National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Paul G Williams
- Center of Chest Disease and Critical Care, Milpark Hospital, Johannesburg, South Africa
| | - Rogerio Souza
- Pulmonary Circulation Unit, Pulmonary Division, Heart Institute (InCor), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
58
|
Whipple NS, Naik RJ, Kang G, Moen J, Govindaswamy SD, Fowler JA, Dowdy J, Penkert R, Joshi VM, Hankins JS. Ventricular global longitudinal strain is altered in children with sickle cell disease. Br J Haematol 2018; 183:796-806. [PMID: 30450553 DOI: 10.1111/bjh.15607] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/08/2018] [Indexed: 12/21/2022]
Abstract
Cardiac disease is the primary cause of death in sickle cell disease (SCD). Right and left ventricular global longitudinal strain (RVGLS, LVGLS) are early markers of systolic dysfunction but are not well investigated among children with SCD. One hundred and forty-three patients with HbSS or HbSβ0 -thalassaemia (median age 11 years, range 5-19 years) and 71 controls matched for age and sex were compared. RVGLS and LVGLS were measured and compared with conventional measures of echocardiography and markers of haemolysis and inflammation. RVGLS was higher in children with SCD than in controls (-25·72% ± 3·45% vs. -24·54% ± 2·41%, P = 0·005); LVGLS was not different. RVGLS decreased with older age in children with SCD (ρ = 0·338, P < 0·001) but not among controls. Decreased RVGLS was associated with increased left atrial end diastolic volume (ρ = 0·181, P = 0·04); RVGLS increased with cardiac output (r = -0·279, P = 0·01). RVGLS and LVGLS were not associated with disease-modifying therapies, degree of anaemia or haemolysis markers. Elevated RVGLS may indicate an early RV compensatory mechanism in response to upstream myocardial insults and elevated cardiac output. Global longitudinal strain may serve as an early marker of altered myocardial function in children with SCD.
Collapse
Affiliation(s)
- Nicholas S Whipple
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ronak J Naik
- Division of Pediatric Cardiology, Le Bonheur Children's Hospital, Memphis, TN, USA.,Cardiopulmonary Services, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Joseph Moen
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - James A Fowler
- Cardiopulmonary Services, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jolanta Dowdy
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rhiannon Penkert
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Vijaya M Joshi
- Division of Pediatric Cardiology, Le Bonheur Children's Hospital, Memphis, TN, USA.,Cardiopulmonary Services, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jane S Hankins
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
59
|
Ezekekwu CA, Kotila TR, Akingbola TS, Lettre G, Gordeuk VR, Cooper RS, DeBaun MR, Inusa B, Tayo BO. Sickle Cell Disease Clinical Trials and Phenotypes. ACTA ACUST UNITED AC 2018; 6:259. [PMID: 30410998 PMCID: PMC6219473 DOI: 10.4172/2329-891x.1000259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Sickle cell disease, one of the world’s most common genetic disorders is prevalent in sub-Saharan Africa. The trans-Atlantic slave trade accounted for the gene movement from Africa to the Caribbean and United States of America and lately, migration has resulted in the introduction of the gene to the United Kingdom and other parts of Europe. Different haplotypes exist, however the differences in these haplotypes are not sufficient to explain the different clinical variations within the same region or different settings.
Collapse
Affiliation(s)
- Chinedu A Ezekekwu
- Department of Hematology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Taiwo R Kotila
- Department of Hematology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Titilola S Akingbola
- Department of Hematology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Victor R Gordeuk
- Division of Hematology & Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Richard S Cooper
- Department of Public Health Sciences, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | | | - Baba Inusa
- Division of Hematology/Oncology, Department of Pediatrics, Vanderbilt Meharry, Center of Excellence in Sickle Cell Disease, Children's Hospital at Vanderbilt, Nashville, USA
| | - Bamidele O Tayo
- Department of Public Health Sciences, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | | |
Collapse
|
60
|
Di Maggio R, Conrey A, Taylor TN, Lederman RJ, Rogers T, Nichols J, Bouges S, Minniti CP, Kato GJ, Shet AS, Hsieh MM. Sickle related events following cardiac catheterisation: risk implication for other invasive procedures. Br J Haematol 2018; 185:778-780. [PMID: 30351508 DOI: 10.1111/bjh.15618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Rosario Di Maggio
- Sickle Cell Branch, NHLBI/NIH, Bethesda, MD, USA.,Campus of Haematology Franco and Piera Cutino, AOOR Villa Sofia-V. Cervello, Palermo, Italy
| | - Anna Conrey
- Sickle Cell Branch, NHLBI/NIH, Bethesda, MD, USA
| | | | | | - Toby Rogers
- Cardiology Branch, NHLBI/NIH, Bethesda, MD, USA
| | - Jim Nichols
- Sickle Cell Branch, NHLBI/NIH, Bethesda, MD, USA
| | - Shenikqua Bouges
- Department of Medicine, University of South Alabama, Mobile, AL, USA
| | - Caterina P Minniti
- Sickle Cell Branch, NHLBI/NIH, Bethesda, MD, USA.,Montefiore Medical Center, Bronx, NY, USA
| | - Gregory J Kato
- Sickle Cell Branch, NHLBI/NIH, Bethesda, MD, USA.,University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Arun S Shet
- Sickle Cell Branch, NHLBI/NIH, Bethesda, MD, USA
| | | |
Collapse
|
61
|
Sundd P, Gladwin MT, Novelli EM. Pathophysiology of Sickle Cell Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:263-292. [PMID: 30332562 DOI: 10.1146/annurev-pathmechdis-012418-012838] [Citation(s) in RCA: 400] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Since the discovery of sickle cell disease (SCD) in 1910, enormous strides have been made in the elucidation of the pathogenesis of its protean complications, which has inspired recent advances in targeted molecular therapies. In SCD, a single amino acid substitution in the β-globin chain leads to polymerization of mutant hemoglobin S, impairing erythrocyte rheology and survival. Clinically, erythrocyte abnormalities in SCD manifest in hemolytic anemia and cycles of microvascular vaso-occlusion leading to end-organ ischemia-reperfusion injury and infarction. Vaso-occlusive events and intravascular hemolysis promote inflammation and redox instability that lead to progressive small- and large-vessel vasculopathy. Based on current evidence, the pathobiology of SCD is considered to be a vicious cycle of four major processes, all the subject of active study and novel therapeutic targeting: ( a) hemoglobin S polymerization, ( b) impaired biorheology and increased adhesion-mediated vaso-occlusion, ( c) hemolysis-mediated endothelial dysfunction, and ( d) concerted activation of sterile inflammation (Toll-like receptor 4- and inflammasome-dependent innate immune pathways). These molecular, cellular, and biophysical processes synergize to promote acute and chronic pain and end-organ injury and failure in SCD. This review provides an exhaustive overview of the current understanding of the molecular pathophysiology of SCD, how this pathophysiology contributes to complications of the central nervous and cardiopulmonary systems, and how this knowledge is being harnessed to develop current and potential therapies.
Collapse
Affiliation(s)
- Prithu Sundd
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA; .,Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Mark T Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA; .,Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Enrico M Novelli
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| |
Collapse
|
62
|
Turpin M, Chantalat-Auger C, Parent F, Driss F, Lionnet F, Habibi A, Maître B, Huertas A, Jaïs X, Weatherald J, Montani D, Sitbon O, Simonneau G, Galactéros F, Humbert M, Bartolucci P, Savale L. Chronic blood exchange transfusions in the management of pre-capillary pulmonary hypertension complicating sickle cell disease. Eur Respir J 2018; 52:52/4/1800272. [PMID: 30305330 DOI: 10.1183/13993003.00272-2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/10/2018] [Indexed: 01/18/2023]
Abstract
The long-term effects of chronic blood exchange transfusions (BETs) on pre-capillary pulmonary hypertension complicating sickle cell disease (SCD) are unknown.13 homozygous SS SCD patients suffering from pre-capillary pulmonary hypertension and treated by chronic BETs were evaluated retrospectively. Assessments included haemodynamics, New York Heart Association Functional Class (NYHA FC), 6-min walk distance (6MWD) and blood tests.Before initiating BETs, all patients were NYHA FC III or IV, median (range) 6MWD was 223 (0-501) m and median (range) pulmonary vascular resistance (PVR) was 3.7 (2-12.5) Wood Units. After a median number of 4 BET sessions, all patients had improved to NYHA FC II or III. Significant improvements in haemodynamics were observed, including a decrease in PVR (p=0.01). There was a trend to higher 6MWD (p=0.09). Median (range) follow-up time after initiation of BETs was 25 (6-53) months. During this period, two patients decided to stop BETs. One of them died from acute right heart failure and the other experienced worsening pulmonary hypertension. Two other patients died during follow-up at 25 and 54 months after BET initiation.Chronic BETs may be a potential therapeutic option in pre-capillary pulmonary hypertension complicating SCD, leading to significant clinical and haemodynamic improvements. These data must be confirmed in a prospective study.
Collapse
Affiliation(s)
- Matthieu Turpin
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Christelle Chantalat-Auger
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Médecine Interne, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,These two authors contributed equally to this work
| | - Florence Parent
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France.,These two authors contributed equally to this work
| | - Françoise Driss
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Médecine Interne, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - François Lionnet
- AP-HP, Service de Médecine Interne, Hôpital Tenon, Paris, France
| | - Anoosha Habibi
- AP-HP, Unité des Maladies Génétiques du Globule Rouge, Centre de Référence des Pathologies du Globule Rouge, Hôpitaux Universitaire Henri Mondor, Créteil, France.,UPEC, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, DHU A-TVB, Créteil, France
| | - Bernard Maître
- AP-HP, Unité de Pneumologie, Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - Alice Huertas
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Xavier Jaïs
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Jason Weatherald
- Division of Respirology, Dept of Medicine, University of Calgary, Calgary, AB, Canada.,Libin Cardiovascular Institute of Alberta, Calgary, AB, Canada
| | - David Montani
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Olivier Sitbon
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Gérald Simonneau
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Fréderic Galactéros
- AP-HP, Unité des Maladies Génétiques du Globule Rouge, Centre de Référence des Pathologies du Globule Rouge, Hôpitaux Universitaire Henri Mondor, Créteil, France.,UPEC, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, DHU A-TVB, Créteil, France
| | - Marc Humbert
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Pablo Bartolucci
- AP-HP, Unité des Maladies Génétiques du Globule Rouge, Centre de Référence des Pathologies du Globule Rouge, Hôpitaux Universitaire Henri Mondor, Créteil, France.,UPEC, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, DHU A-TVB, Créteil, France
| | - Laurent Savale
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| |
Collapse
|
63
|
How I treat the older adult with sickle cell disease. Blood 2018; 132:1750-1760. [PMID: 30206116 DOI: 10.1182/blood-2018-03-818161] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/21/2018] [Indexed: 12/31/2022] Open
Abstract
With increasing survival, cumulative complications of sickle cell disease (SCD), which develop insidiously over time, are becoming more apparent and common in older patients, particularly those in their fifth decade and beyond. The older patient is also more likely to develop other age-related nonsickle conditions that interact and add to the disease morbidity. A common misconception is that any symptom in a SCD patient is attributable to their SCD and this may lead to delays in diagnosis and appropriate intervention. We recommend regular comprehensive reviews and monitoring for early signs of organ damage and a low threshold for the use of hydroxyurea and blood transfusions as preventative measures for end-organ disease. Treatable comorbidities and acute deterioration should be managed aggressively. Although the primary goal in management of the older adult with SCD is improving anemia and minimizing organ damage, the time has come for us to be more proactive in considering curative therapies previously offered to the younger patient. Curative or experimental interventions should be discussed early, before complications render the patients ineligible for these treatments.
Collapse
|
64
|
How I treat hypoxia in adults with hemoglobinopathies and hemolytic disorders. Blood 2018; 132:1770-1780. [PMID: 30206115 DOI: 10.1182/blood-2018-03-818195] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 06/13/2018] [Indexed: 01/19/2023] Open
Abstract
Hemoglobinopathies are caused by genetic mutations that result in abnormal hemoglobin molecules, resulting in hemolytic anemia. Chronic complications involving the lung parenchyma, vasculature, and cardiac function in hemoglobinopathies result in impaired gas exchange, resulting in tissue hypoxia. Hypoxia is defined as the deficiency in the amount of oxygen reaching the tissues of the body and is prevalent in patients with hemoglobinopathies, and its cause is often multifactorial. Chronic hypoxia in hemoglobinopathies is often a sign of disease severity and is associated with increased morbidity and mortality. Therefore, a thorough understanding of the pathophysiology of hypoxia in these disease processes is important in order to appropriately treat the underlying cause and prevent complications. In this article, we discuss management of hypoxia based on three different cases: sickle cell disease, β-thalassemia, and hereditary spherocytosis. These cases are used to review the current understanding of the disease pathophysiology, demonstrate the importance of a thorough clinical history and physical examination, explore diagnostic pathways, and review the current management.
Collapse
|
65
|
Musa BM, Odoh CN, Galadanci NA, Saidu H, Aliyu MH. Lower than expected elevated tricuspid regurgitant jet velocity in adults with sickle cell disease in Nigeria. Int Health 2018; 10:356-362. [PMID: 29438485 DOI: 10.1093/inthealth/ihx074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 12/13/2017] [Indexed: 11/15/2022] Open
Abstract
Background Cardiopulmonary disease is a major cause of morbidity and mortality in persons with sickle cell disease (SCD). Tricuspid regurgitant jet velocity (TRJV) and predicted forced expiratory volume in 1 s (FEV1%) predicted are independently associated with death in SCD. The goal of this study was to determine the prevalence of elevated TRJV and the association, if any, between TRJV and FEV1% predicted among persons with sickle cell anaemia (SCA) in Nigeria. Methods Using a cross-sectional design, we enrolled 100 adult Nigerians (≥15 y) with SCA. We screened participants using Doppler echocardiogram to determine their TRJV and assessed their lung function with spirometry. Results The prevalence of elevated TRJV was 6%, with 74% of participants having low FEV1% predicted (<70%). TRJV was negatively correlated with FEV1%, but this finding was not statistically significant (Spearman's ρ=-0.0263, p=0.8058). Conclusions We found a low prevalence of elevated TRJV and a trend in association between TRJV and FEV1% predicted in Nigerian adults with SCA. Our findings underscore the need to explore further the relationship between SCD and cardiopulmonary disease in adults.
Collapse
Affiliation(s)
- Baba Maiyaki Musa
- Department of Medicine, Bayero University and Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Chisom N Odoh
- Department of Epidemiology and Population Health, University of Louisville, Louisville, Kentucky, USA
| | - Najibah A Galadanci
- Department of Hematology, Bayero University and Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Hadiza Saidu
- Department of Medicine, Bayero University and Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Muktar H Aliyu
- Vanderbilt Institute for Global Health, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Health Policy, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
66
|
Abstract
In the 100 years since sickle cell anemia (SCA) was first described in the medical literature, studies of its molecular and pathophysiological basis have been at the vanguard of scientific discovery. By contrast, the translation of such knowledge into treatments that improve the lives of those affected has been much too slow. Recent years, however, have seen major advances on several fronts. A more detailed understanding of the switch from fetal to adult hemoglobin and the identification of regulators such as BCL11A provide hope that these findings will be translated into genomic-based approaches to the therapeutic reactivation of hemoglobin F production in patients with SCA. Meanwhile, an unprecedented number of new drugs aimed at both the treatment and prevention of end-organ damage are now in the pipeline, outcomes from potentially curative treatments such as allogeneic hematopoietic stem cell transplantation are improving, and great strides are being made in gene therapy, where methods employing both antisickling β-globin lentiviral vectors and gene editing are now entering clinical trials. Encouragingly, after a century of neglect, the profile of the vast majority of those with SCA in Africa and India is also finally improving.
Collapse
Affiliation(s)
- Thomas N Williams
- Department of Epidemiology and Demography, KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Medicine, Imperial College London, London W2 1NY, United Kingdom;
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1589, USA;
| |
Collapse
|
67
|
Vanderpool RR, Naeije R. Hematocrit-corrected Pulmonary Vascular Resistance. Am J Respir Crit Care Med 2018. [DOI: 10.1164/rccm.201801-0081pp] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Rebecca R. Vanderpool
- Department of Translational and Regenerative Medicine, University of Arizona, Tucson, Arizona; and
| | - Robert Naeije
- Department of Cardiology, Erasmus University Hospital, Brussels, Belgium
| |
Collapse
|
68
|
Bilan VP, Schneider F, Novelli EM, Kelley EE, Shiva S, Gladwin MT, Jackson EK, Tofovic SP. Experimental intravascular hemolysis induces hemodynamic and pathological pulmonary hypertension: association with accelerated purine metabolism. Pulm Circ 2018; 8:2045894018791557. [PMID: 30003836 PMCID: PMC6080084 DOI: 10.1177/2045894018791557] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Pulmonary hypertension (PH) is emerging as a serious complication associated with
hemolytic disorders, and plexiform lesions (PXL) have been reported in patients
with sickle cell disease (SCD). We hypothesized that repetitive hemolysis per se
induces PH and angioproliferative vasculopathy and evaluated a new mechanism for
hemolysis-associated PH (HA-PH) that involves the release of adenosine deaminase
(ADA) and purine nucleoside phosphorylase (PNP) from erythrocytes. In healthy
rats, repetitive administration of hemolyzed autologous blood (HAB) for 10 days
produced reversible pulmonary parenchymal injury and vascular remodeling and PH.
Moreover, the combination of a single dose of Sugen-5416 (SU, 200 mg/kg) and
10-day HAB treatment resulted in severe and progressive obliterative PH and
formation of PXL (Day 26, right ventricular peak systolic pressure (mmHg):
26.1 ± 1.1, 41.5 ± 0.5 and 85.1 ± 5.9 in untreated, HAB treated and SU+HAB
treated rats, respectively). In rats, repetitive administration of HAB increased
plasma ADA activity and reduced urinary adenosine levels. Similarly, SCD
patients had higher plasma ADA and PNP activity and accelerated adenosine,
inosine, and guanosine metabolism than healthy controls. Our study provides
evidence that hemolysis per se leads to the development of angioproliferative
PH. We also report the development of a rat model of HA-PH that closely mimics
pulmonary vasculopathy seen in patients with HA-PH. Finally, this study suggests
that in hemolytic diseases released ADA and PNP may increase the risk of PH,
likely by abolishing the vasoprotective effects of adenosine, inosine and
guanosine. Further characterization of this new rat model of hemolysis-induced
angioproliferative PH and additional studies of the role of purines metabolism
in HA-PH are warranted.
Collapse
Affiliation(s)
- Victor P Bilan
- 1 Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,3 Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Frank Schneider
- 4 Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Enrico M Novelli
- 2 Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,3 Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eric E Kelley
- 5 Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Sruti Shiva
- 2 Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,6 Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mark T Gladwin
- 1 Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,3 Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Edwin K Jackson
- 6 Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stevan P Tofovic
- 1 Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,3 Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
69
|
Mehari A, Igbineweka N, Allen D, Nichols J, Thein SL, Weir NA. Abnormal Ventilation-Perfusion Scan Is Associated with Pulmonary Hypertension in Sickle Cell Adults. J Nucl Med 2018; 60:86-92. [PMID: 29880507 DOI: 10.2967/jnumed.118.211466] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/23/2018] [Indexed: 02/02/2023] Open
Abstract
Pulmonary hypertension (PH) in adults with sickle cell disease (SCD) is associated with early mortality. Chronic thromboembolic PH (CTEPH) is an important complication and contributor to PH in SCD but is likely underappreciated. Guidelines recommend ventilation-perfusion (V/Q) scintigraphy as the imaging modality of choice to exclude CTEPH. Data on V/Q scanning are limited in SCD. Our objective was to compare the performance of V/Q scanning with that of CT pulmonary angiography (CTPA) and to report clinical outcomes associated with abnormal V/Q findings. Methods: Laboratory data, echocardiography, 6-min-walk testing, V/Q scanning, CTPA, and right heart catheterization (RHC) were prospectively obtained. High-probability and intermediate-probability V/Q findings were considered to be abnormal. Included for analysis were 142 SCD adults (aged 40.1 ± 13.7 y, 83 women, 87% hemoglobin SS) in a stable state enrolled consecutively between March 13, 2002, and June 8, 2017. Results: V/Q results were abnormal in 65 of 142 patients (45.8%). CTPA was positive for pulmonary embolism in 16 of 60 (26.7%). RHC confirmed PH (mean pulmonary artery pressure ≥ 25 mmHg) in 46 of 64 (71.9%), of whom 34 (73.9%) had abnormal V/Q findings. Among those without PH by RHC (n = 18), 2 of 18 patients had abnormal V/Q findings. Thirty-three patients had a complete dataset (V/Q scanning, CTPA, and RHC); 29 of 33 had abnormal RHC findings, of whom 26 had abnormal V/Q findings, compared with 11 who had abnormal CTPA findings. There was greater concordance between V/Q findings and RHC (κ-value = 0.53; P < 0.001) than between CTPA and RHC (κ-value = 0.13; P = 0.065). The sensitivity and specificity for V/Q scanning was 89.7% and 75.0%, respectively, whereas CTPA had sensitivity of 37.3% and specificity of 100%. Abnormal V/Q finding swere associated with hemodynamic severity (mean pulmonary artery pressure, 35.2 ± 9.6 vs. 26.9 ± 10.5 mm Hg, P = 0.002; transpulmonary gradient, 21.5 ± 9.7 vs. 12.16 ± 11 mmHg, P = 0.005; and pulmonary vascular resistance, 226.5 ± 135 vs. 140.7 ± 123.7 dynes⋅s⋅cm-5, P = 0.013) and exercise capacity (6-min-walk distance, 382.8 ± 122.3 vs. 442.3 ± 110.6 m, P < 0.010). Thirty-four deaths were observed over 15 y. All-cause mortality was higher in the abnormal-V/Q group (21 [61.8%]) than in the normal-V/Q group (13 [38.2%]) (log-rank test, P = 0.006; hazard ratio, 2.54). Conclusion: V/Q scanning is superior to CTPA in detecting thrombotic events in SCD. Abnormal V/Q findings are associated with PH, worse hemodynamics, lower functional capacity, and higher mortality. Despite high sensitivity in detecting CTEPH, V/Q scanning is underutilized. We recommend the use of V/Q scanning in the evaluation of dyspnea in adult SCD patients given the important implications toward management.
Collapse
Affiliation(s)
- Alem Mehari
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland .,Division of Pulmonary Diseases, Howard University College of Medicine, Washington, District of Columbia; and
| | - Norris Igbineweka
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Darlene Allen
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Jim Nichols
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Nargues A Weir
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,Inova Advanced Lung Disease Program, Falls Church, Virginia
| |
Collapse
|
70
|
Abstract
Non-adherence and deformability are the key intrinsic biomechanical features of the red blood cell (RBC), which allow it to tightly squeeze and pass through even the narrowest of microcirculatory networks. Blockage of microcirculatory flow, also known as vaso-occlusion, is a consequence of abnormal cellular adhesion to the vascular endothelium. In sickle cell disease (SCD), an inherited anaemia, even though RBCs have been shown to be heterogeneous in adhesiveness and deformability, this has not been studied in the context of physiologically relevant dynamic shear gradients at the microscale. We developed a microfluidic system that simulates physiologically relevant shear gradients of microcirculatory blood flow at a constant single volumetric flow rate. Using this system, shear dependent adhesion of RBCs from 28 subjects with SCD and from 11 healthy subjects was investigated using vascular endothelial protein functionalized microchannels. We defined a new term, RBC Shear Gradient Microfluidic Adhesion (SiGMA) index to assess shear dependent RBC adhesion in a subject-specific manner. We have shown for the first time that shear dependent adhesion of RBCs is heterogeneous in a microfluidic flow model, which correlates clinically with inflammatory markers and iron overload in subjects with SCD. This study reveals the complex dynamic interactions between RBC-mediated microcirculatory occlusion and clinical outcomes in SCD. These interactions may also be relevant to other microcirculatory disorders and microvascular diseases.
Collapse
Affiliation(s)
- Erdem Kucukal
- Department of Mechanical and Aerospace Engineering, Case Biomanufacturing and Microfabrication Laboratory, Case Western Reserve University, Glennan 616B, 10900 Euclid Ave., Cleveland, OH, USA.
| | - Jane A Little
- Department of Hematology and Oncology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA and Seidman Cancer Center at University Hospitals, Case Medical Center, Cleveland, OH, USA
| | - Umut A Gurkan
- Department of Mechanical and Aerospace Engineering, Case Biomanufacturing and Microfabrication Laboratory, Case Western Reserve University, Glennan 616B, 10900 Euclid Ave., Cleveland, OH, USA. and Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA and Department of Orthopaedics, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
71
|
Haw A, Palevsky HI. Pulmonary hypertension in chronic hemolytic anemias: Pathophysiology and treatment. Respir Med 2018; 137:191-200. [DOI: 10.1016/j.rmed.2018.02.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/31/2018] [Accepted: 02/26/2018] [Indexed: 01/19/2023]
|
72
|
Abstract
Sickle cell disease (SCD) is a monogenetic disorder caused by a mutation in the β-globin gene HBB leading to polymerization of red blood cells causing damage to cell membranes, increasing its rigidity and intravascular hemolysis. Multiple lines of evidence suggest that SCD can be viewed as pan-vasculopathy associated with multiple mechanisms but driven by hemoglobin S polymerization. Here we review the pathophysiology, clinical manifestations and management strategies for cerebrovascular disease, pulmonary hypertension and renal disease associated with SCD. These "vascular phenotypes" reflect the systemic nature of the complications of SCD and are a major threat to the well-being of patients with the disorder.
Collapse
Affiliation(s)
- Ashar Usmani
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA
| | - Roberto F. Machado
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
73
|
Lilje C, Harry J, Gajewski KK, Gardner RV. A modified noninvasive screening protocol for pulmonary hypertension in children with sickle cell disease-Who should be sent for invasive evaluation? Pediatr Blood Cancer 2017; 64. [PMID: 28475298 DOI: 10.1002/pbc.26606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 01/23/2023]
Abstract
BACKGROUND Invasive studies have shown that prevalence and severity of pulmonary hypertension (PH) in patients with sickle cell disease (SCD) tend to be overestimated if based exclusively on Doppler-derived tricuspid regurgitant velocity (TRV) as surrogate noninvasive marker with a cutoff ≥2.5 m/s. OBJECTIVES We aimed to better define a subgroup of pediatric SCD patients who should be sent for invasive evaluation of pulmonary artery pressure (PAP) based on a modified echocardiographic PH screening protocol that implements evidence from Doppler-catheter comparative studies. STUDY DESIGN Charts of 121 pediatric patients with stable SCD were reviewed regarding echocardiographically assessed risk for elevated PAP/PH and associated clinical characteristics. TRV cutoff was refined at ≥2.9 m/s to avoid overestimating the risk for PH. TRV was combined with additional echocardiographic parameters to avoid underestimating the PH risk. RESULTS Ninety-one patients qualified for analysis. Based on our modified echocardiographic protocol, 5.5% of patients qualified for at least moderate risk for elevated PAP (compatible with PH) as opposed to 20.9% if based exclusively on TRV ≥2.5 m/s. These patients were older, homozygous for hemoglobin S (HbSS), and more anemic. No subject had an echocardiographic risk constellation suggesting more than mild PH. CONCLUSIONS Our modified noninvasive screening protocol-if confirmed by invasive studies-may help to better identify a subgroup of pediatric SCD patients in whom evaluation by catheterization appears justified. Unlike estimates based on the conventional protocol, the size of the targeted subgroup compares favorably with catheterization-confirmed PH prevalence rates. Characteristics associated with an increased PH risk were also identified.
Collapse
Affiliation(s)
- Christian Lilje
- Division of Pediatric Cardiology, Department of Pediatrics, Louisiana State University Health Sciences Center/Children's Hospital, New Orleans, Louisiana
| | - Jordan Harry
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Kelly K Gajewski
- Division of Pediatric Cardiology, Department of Pediatrics, Louisiana State University Health Sciences Center/Children's Hospital, New Orleans, Louisiana
| | - Renee V Gardner
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Louisiana State University Health Sciences Center/Children's Hospital, New Orleans, Louisiana
| |
Collapse
|
74
|
Rai P, Niss O, Malik P. A reappraisal of the mechanisms underlying the cardiac complications of sickle cell anemia. Pediatr Blood Cancer 2017; 64. [PMID: 28453224 DOI: 10.1002/pbc.26607] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/07/2017] [Accepted: 03/24/2017] [Indexed: 12/28/2022]
Abstract
Anemia, hemolysis-driven vasculopathy, and intrinsic myocardial injury have been proposed as predisposing factors to cardiac disease in sickle cell anemia (SCA). The individual impact of these mechanisms on the cardiac features of SCA and the way they influence complications such as sudden death and dysrhythmias have been unclear. Recent findings of an acquired restrictive SCA-related cardiomyopathy, driven by myocardial fibrosis, may explain some of these cardiac features. Given the complexity of cardiac pathology in SCA, using additional parameters to tricuspid regurgitant jet velocity (left atrial volume, diastolic parameters, NT-proBNP) may improve the accuracy of noninvasive screening for cardiopulmonary complications in SCA.
Collapse
Affiliation(s)
- Parul Rai
- Division of Experimental Hematology & Cancer Biology and Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Omar Niss
- Division of Experimental Hematology & Cancer Biology and Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Punam Malik
- Division of Experimental Hematology & Cancer Biology and Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
75
|
Medical Management of Pulmonary Hypertension with Unclear and/or Multifactorial Mechanisms (Group 5): Is There a Role for Pulmonary Arterial Hypertension Medications? Curr Hypertens Rep 2017; 19:86. [PMID: 29046979 DOI: 10.1007/s11906-017-0783-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review was to outline the mechanisms and to review recent literature on pulmonary arterial hypertension (PAH) medications in group 5 pulmonary hypertension (PH). RECENT FINDINGS The first steps in management are to understand the mechanisms and hemodynamic profile and to exclude chronic thromboembolic disease. Recent studies in the past 5 years have found that PAH medications may improve hemodynamics in patients with pre-capillary pulmonary hypertension due to sarcoidosis, pulmonary Langerhans cell histiocytosis, lymphangioleiomyomatosis, and myeloproliferative disorders with dasatinib-induced PH. Improvements in exercise capacity are uncommon, and no survival benefit has been demonstrated. There is a risk of pulmonary edema in patients with pulmonary venous involvement or fibrosing mediastinitis when treated with PAH therapies. There is limited evidence supporting the use of PAH medications in group 5 patients, and they may be harmful in certain cases. In most patients with group 5 PH, treatment should be directed to the underlying disease with PAH therapies reserved for patients with severe pre-capillary PH.
Collapse
|
76
|
Real-life experience with hydroxyurea in sickle cell disease: A multicenter study in a cohort of patients with heterogeneous descent. Blood Cells Mol Dis 2017; 69:82-89. [PMID: 29107441 DOI: 10.1016/j.bcmd.2017.08.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022]
Abstract
We conducted the first nation-wide cohort study of sickle cell disease (SCD) in Italy, a Southern European country exposed to intense recent flux migration from endemic areas for SCD. We evaluate the impact of hydroxyurea on a total of 652 pediatric and adult patients from 33 Reference Centers for SCD (mean age 24.5±15years, 51.4% males). Hydroxyurea median treatment duration was 7years (range: <1year to 29years) at a mean therapeutic dose of 18±4.7mg/kg/day. Hydroxyurea was associated with a significant increase in mean total and fetal hemoglobin and a significant decrease in mean hemoglobin S, white blood and platelet counts, and lactate dehydrogenase levels. Hydroxyurea was associated with a significant reduction in the incidence of acute chest syndrome (-29.3%, p<0.001), vaso-occlusive crisis (-34.1%, p<0.001), hospitalization (-53.2%, p<0.001), and bone necrosis (-6.9%, p<0.001). New silent cerebral infarction (SCI) occurred during treatment (+42.4%, p<0.001) but not stroke (+0.5%, p=0.572). These observations were generally consistent upon stratification for age, descent (Caucasian or African), genotype (βS/βS, βS/β0 or βS/β+) and duration of treatment (< or ≥10years). There were no new safety concerns observed compared to those commonly reported in the literature. Our study, conducted on a large population of patients with different descent and compound state supports the benefits of hydroxyurea therapy as a treatment option. Registered at clinical trials.gov (NCT02709681).
Collapse
|
77
|
Shilo NR, Morris CR. Pathways to pulmonary hypertension in sickle cell disease: the search for prevention and early intervention. Expert Rev Hematol 2017; 10:875-890. [PMID: 28817980 DOI: 10.1080/17474086.2017.1364989] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Pulmonary hypertension (PH) develops in a significant number of patients with sickle cell disease (SCD), resulting in increased morbidity and mortality. This review focuses on PH pathophysiology, risk stratification, and new recommendations for screening and treatment for patients with SCD. Areas covered: An extensive PubMed literature search was performed. While the pathophysiology of PH in SCD is yet to be fully deciphered, it is known that the etiology is multifactorial; hemolysis, hypercoagulability, hypoxemia, ischemic-reperfusion injury, oxidative stress, and genetic susceptibility all contribute in varying degrees to endothelial dysfunction. Hemolysis, in particular, seems to play a key role by inciting an imbalance in the regulatory axis of nitric oxide and arginine metabolism. Systematic risk stratification starting in childhood based on clinical features and biomarkers that enable early detection is necessary. Multi-faceted, targeted interventions, before irreversible vasculopathy develops, will allow for improved patient outcomes and life expectancy. Expert commentary: Despite progress in our understanding of PH in SCD, clinically proven therapies remain elusive and additional controlled clinical trials are needed. Prevention of disease starts in childhood, a critical window for intervention. Given the complex and multifactorial nature of SCD, patients will ultimately benefit from combination therapies that simultaneously targets multiple mechanisms.
Collapse
Affiliation(s)
- Natalie R Shilo
- a Department of Pediatrics, Division of Pulmonary Medicine , University of Connecticut Heath Center , Farmington , CT , USA
| | - Claudia R Morris
- b Department of Pediatrics, Division of Pediatric Emergency Medicine, Emory-Children's Center for Cystic Fibrosis and Airways Disease Research , Emory University School of Medicine , Atlanta , GA , USA
| |
Collapse
|
78
|
Goldberg AB, Mazur W, Kalra DK. Pulmonary hypertension: diagnosis, imaging techniques, and novel therapies. Cardiovasc Diagn Ther 2017; 7:405-417. [PMID: 28890877 DOI: 10.21037/cdt.2017.04.11] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Pulmonary hypertension (PH), defined as the elevation of mean pulmonary arterial pressure (mPAP) above 25 mmHg, has numerous causes, which the World Health Organization (WHO) has divided into five distinct categories based upon the underlying mechanism of action. This review will explore the pathophysiology, diagnostic testing, and treatment of PH. Identification of PH depends on a strong clinical suspicion, a detailed history, and a thorough physical exam. We review the evidence supporting experimental and clinical laboratory parameters for diagnosis and monitoring of PH. Transthoracic echocardiogram (TTE) is the initial screening test of choice. This review will detail specific echocardiographic techniques for the assessment and classification of PH. Furthermore, the importance of advanced imaging, including computed tomography (CT) and magnetic resonance imagining (MRI) is explored. New developments in pharmacology, percutaneous intervention, and surgical approaches are summarized. Finally, we will address the tools available to predict morbidity and mortality.
Collapse
Affiliation(s)
- Alan B Goldberg
- Department of Internal Medicine, Division of Cardiology, Rush University Medical Center, Chicago, Illinois, USA
| | | | - Dinesh K Kalra
- Department of Internal Medicine, Division of Cardiology, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
79
|
Lopes AJ, Marinho CL, Alves UD, Gonçalves CEA, Silva PO, Botelho EC, Bedirian R, Soares AR, Maioli MCP. Relationship between ventilation heterogeneity and exercise intolerance in adults with sickle cell anemia. ACTA ACUST UNITED AC 2017; 50:e6512. [PMID: 28746470 PMCID: PMC5520223 DOI: 10.1590/1414-431x20176512] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/09/2017] [Indexed: 12/18/2022]
Abstract
Sickle cell anemia (SCA) causes dysfunction of multiple organs, with pulmonary involvement as a major cause of mortality. Recently, there has been growing interest in the nitrogen single-breath washout (N2SBW) test, which is able to detect ventilation heterogeneity and small airway disease when the results of other pulmonary function tests (PFTs) are still normal. Thus, the objectives of the present study were to assess the heterogeneity in the ventilation distribution in adults with SCA and to determine the association between the ventilation distribution and the clinical, cardiovascular, and radiological findings. This cross-sectional study included 38 adults with SCA who underwent PFTs, echocardiography, computed tomography (CT), and 6-min walk test. To evaluate the ventilation heterogeneity, the patients were categorized according to the phase III slope of the N2SBW (SIIIN2). Compared with adults with lower SIIIN2 values, adults with higher SIIIN2 values showed lower hemoglobin levels (P=0.048), a history of acute chest syndrome (P=0.001), an elevated tricuspid regurgitation velocity (P=0.039), predominance of a reticular pattern in the CT (P=0.002), a shorter 6-min walking distance (6MWD) (P=0.002), and lower peripheral oxygen saturation (SpO2) after exercise (P=0.03). SIIIN2 values correlated significantly with hemoglobin (rs=-0.344; P=0.034), forced vital capacity (rs=-0.671; P<0.0001), diffusing capacity for carbon monoxide (rs=-0.376; P=0.019), 6MWD (rs=-0.554; P=0.0003), and SpO2 after exercise (P=0.040). Heterogeneity in the ventilation distribution is one of the most common pulmonary dysfunctions in adults with SCA. Moreover, relationships exist between ventilation heterogeneity, worsening of pulmonary structural damage, and reduced tolerance for exercise.
Collapse
Affiliation(s)
- A J Lopes
- Programa de Pós-Graduação em Ciências da Reabilitação, Centro Universitário Augusto Motta, Rio de Janeiro, RJ, Brasil.,Programa de Pós-Graduação em Ciências Médicas, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - C L Marinho
- Programa de Pós-Graduação em Fisiopatologia Clínica e Experimental, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - U D Alves
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - C E A Gonçalves
- Programa de Pós-Graduação em Ciências da Reabilitação, Centro Universitário Augusto Motta, Rio de Janeiro, RJ, Brasil
| | - P O Silva
- Programa de Pós-Graduação em Ciências da Reabilitação, Centro Universitário Augusto Motta, Rio de Janeiro, RJ, Brasil
| | - E C Botelho
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - R Bedirian
- Disciplina de Clínica Médica, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - A R Soares
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil.,Disciplina de Hematologia e Hemoterapia, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - M C P Maioli
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil.,Disciplina de Hematologia e Hemoterapia, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
80
|
Kang BY, Park K, Kleinhenz JM, Murphy TC, Sutliff RL, Archer D, Hart CM. Peroxisome Proliferator-Activated Receptor γ Regulates the V-Ets Avian Erythroblastosis Virus E26 Oncogene Homolog 1/microRNA-27a Axis to Reduce Endothelin-1 and Endothelial Dysfunction in the Sickle Cell Mouse Lung. Am J Respir Cell Mol Biol 2017; 56:131-144. [PMID: 27612006 DOI: 10.1165/rcmb.2016-0166oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pulmonary hypertension (PH), a serious complication of sickle cell disease (SCD), causes significant morbidity and mortality. Although a recent study determined that hemin release during hemolysis triggers endothelial dysfunction in SCD, the pathogenesis of SCD-PH remains incompletely defined. This study examines peroxisome proliferator-activated receptor γ (PPARγ) regulation in SCD-PH and endothelial dysfunction. PH and right ventricular hypertrophy were studied in Townes humanized sickle cell (SS) and littermate control (AA) mice. In parallel studies, SS or AA mice were gavaged with the PPARγ agonist, rosiglitazone (RSG), 10 mg/kg/day, or vehicle for 10 days. In vitro, human pulmonary artery endothelial cells (HPAECs) were treated with vehicle or hemin for 72 hours, and selected HPAECs were treated with RSG. SS mice developed PH and right ventricular hypertrophy associated with reduced lung levels of PPARγ and increased levels of microRNA-27a (miR-27a), v-ets avian erythroblastosis virus E26 oncogene homolog 1 (ETS1), endothelin-1 (ET-1), and markers of endothelial dysfunction (platelet/endothelial cell adhesion molecule 1 and E selectin). HPAECs treated with hemin had increased ETS1, miR-27a, ET-1, and endothelial dysfunction and decreased PPARγ levels. These derangements were attenuated by ETS1 knockdown, inhibition of miR-27a, or PPARγ overexpression. In SS mouse lung or in hemin-treated HPAECs, activation of PPARγ with RSG attenuated reductions in PPARγ and increases in miR-27a, ET-1, and markers of endothelial dysfunction. In SCD-PH pathogenesis, ETS1 stimulates increases in miR-27a levels that reduce PPARγ and increase ET-1 and endothelial dysfunction. PPARγ activation attenuated SCD-associated signaling derangements, suggesting a novel therapeutic approach to attenuate SCD-PH pathogenesis.
Collapse
Affiliation(s)
- Bum-Yong Kang
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Kathy Park
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Jennifer M Kleinhenz
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Tamara C Murphy
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Roy L Sutliff
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - David Archer
- 2 Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - C Michael Hart
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| |
Collapse
|
81
|
Amadi VN, Balogun MO, Akinola NO, Adebayo RA, Akintomide AO. Pulmonary hypertension in Nigerian adults with sickle cell anemia. Vasc Health Risk Manag 2017; 13:153-160. [PMID: 28507438 PMCID: PMC5428794 DOI: 10.2147/vhrm.s92799] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background Sickle cell anemia (SCA) is the commonest hemoglobinopathy and is associated with high morbidity and mortality. Pulmonary hypertension (PH) is reported to play a significant role in this regard. There is very limited literature on PH in SCA in Nigeria. Objectives The objectives of this study were to determine the prevalence of Doppler-derived PH in SCA, assess its influence on exercise capacity, and determine the correlates and predictors of measures of estimated pulmonary pressure. Methods A total of 92 SCA subjects had echocardiography and 6-minute self-paced walking exercise. PH was diagnosed by Doppler echocardiography on finding a tricuspid regurgitant velocity (TRV) of ≥2.5 m/s. The pulmonary flow profile was also assessed to estimate mean pulmonary arterial pressure (MPAP). Results Doppler-derived PH was detected in 23.9% of adults with SCA. The 6-minute walking distance (6MWD) was significantly lower in SCA adults with PH than in those without PH (380.33 ± 63.17 m vs 474.28 ± 76.74 m; p = 0.014). TRV and estimated MPAP had a significant inverse correlation with the 6MWD (r = −0.442; p < 0.001 and r = −0.571; p < 0.001, respectively). Conclusion PH as derived by Doppler is common in Nigerian adults with SCA and has a significantly negative influence on exercise capacity. Screening for PH should be encouraged to optimize management and thus improve their quality of life and life expectancy.
Collapse
Affiliation(s)
- Valentine N Amadi
- Department of Internal Medicine, Federal Medical Centre, Asaba, Delta State
| | | | - Norah O Akinola
- Department of Haematology and Blood Transfusion, Obafemi Awolowo University Teaching Hospitals Complex, Ile-Ife, Osun State, Nigeria
| | | | | |
Collapse
|
82
|
Freitas CSG, Baldi BG, Jardim C, Araujo MS, Sobral JB, Heiden GI, Kairalla RA, Souza R, Carvalho CRR. Pulmonary hypertension in lymphangioleiomyomatosis: prevalence, severity and the role of carbon monoxide diffusion capacity as a screening method. Orphanet J Rare Dis 2017; 12:74. [PMID: 28427470 PMCID: PMC5399314 DOI: 10.1186/s13023-017-0626-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/05/2017] [Indexed: 01/23/2023] Open
Abstract
Background Lymphangioleiomyomatosis (LAM) is included within group 5 of the current PH classification (unclear multifactorial mechanisms). However, data regarding the occurrence of PH in LAM are scarce. The aims of the study were to describe the prevalence and characteristics of PH in a large cohort of LAM patients with different levels of severity, and to evaluate the role of echocardiography and carbon monoxide diffusion capacity (DLCO) as screening methods for PH in LAM. Methods One hundred five LAM patients underwent transthoracic echocardiography, pulmonary function tests (PFTs) and 6-min walk test (6MWT). Patients with a suspicion of PH on echocardiography, defined by the presence of estimated systolic pulmonary artery pressure (PAP) over 35 mmHg or PFT showing DLco below 40% of the predicted value, underwent right heart catheterisation to confirm the diagnosis of PH. Results Eight patients (7.6%) had PH confirmed on right heart catheterisation, six patients (5.7%) had a pre-capillary pattern and two patients (1.9%) had a post-capillary profile. Only one patient (1%) had mean PAP over 35 mmHg. Patients with PH had lower FEV1 and DLCO in PFTs and greater oxygen desaturation and dyspnea intensity during 6MWT compared with those without PH. In 63% of the patients with confirmed PH, the right heart catheterisation was performed based only on DLCO result. Conclusions The prevalence of PH is low in LAM patients. Pulmonary hypertension in LAM is typically mild and significantly associated with pulmonary parenchymal involvement. Carbon monoxide diffusion capacity significantly improved the identification of PH in LAM patients.
Collapse
Affiliation(s)
- Carolina S G Freitas
- Pulmonary Division, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 44, 5° andar - sala 1, São Paulo, 05403-900, Brazil
| | - Bruno G Baldi
- Pulmonary Division, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 44, 5° andar - sala 1, São Paulo, 05403-900, Brazil.
| | - Carlos Jardim
- Pulmonary Division, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 44, 5° andar - sala 1, São Paulo, 05403-900, Brazil
| | - Mariana S Araujo
- Pulmonology Department, Federal University of Paraná, Curitiba, Brazil
| | - Juliana Barbosa Sobral
- Echocardiography Laboratory, Radiology Institute (InRad), University of São Paulo Medical School, São Paulo, Brazil
| | - Gláucia I Heiden
- Pulmonary Division, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 44, 5° andar - sala 1, São Paulo, 05403-900, Brazil
| | - Ronaldo A Kairalla
- Pulmonary Division, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 44, 5° andar - sala 1, São Paulo, 05403-900, Brazil
| | - Rogério Souza
- Pulmonary Division, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 44, 5° andar - sala 1, São Paulo, 05403-900, Brazil
| | - Carlos R R Carvalho
- Pulmonary Division, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 44, 5° andar - sala 1, São Paulo, 05403-900, Brazil
| |
Collapse
|
83
|
Koudelka A, Ambrozova G, Klinke A, Fidlerova T, Martiskova H, Kuchta R, Rudolph TK, Kadlec J, Kuchtova Z, Woodcock SR, Freeman BA, Kubala L, Pekarova M. Nitro-Oleic Acid Prevents Hypoxia- and Asymmetric Dimethylarginine-Induced Pulmonary Endothelial Dysfunction. Cardiovasc Drugs Ther 2017; 30:579-586. [PMID: 27858190 DOI: 10.1007/s10557-016-6700-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE Pulmonary hypertension (PH) represents a serious health complication accompanied with hypoxic conditions, elevated levels of asymmetric dimethylarginine (ADMA), and overall dysfunction of pulmonary vascular endothelium. Since the prevention strategies for treatment of PH remain largely unknown, our study aimed to explore the effect of nitro-oleic acid (OA-NO2), an exemplary nitro-fatty acid (NO2-FA), in human pulmonary artery endothelial cells (HPAEC) under the influence of hypoxia or ADMA. METHODS HPAEC were treated with OA-NO2 in the absence or presence of hypoxia and ADMA. The production of nitric oxide (NO) and interleukin-6 (IL-6) was monitored using the Griess method and ELISA, respectively. The expression or activation of different proteins (signal transducer and activator of transcription 3, STAT3; hypoxia inducible factor 1α, HIF-1α; endothelial nitric oxide synthase, eNOS; intercellular adhesion molecule-1, ICAM-1) was assessed by the Western blot technique. RESULTS We discovered that OA-NO2 prevents development of endothelial dysfunction induced by either hypoxia or ADMA. OA-NO2 preserves normal cellular functions in HPAEC by increasing NO production and eNOS expression. Additionally, OA-NO2 inhibits IL-6 production as well as ICAM-1 expression, elevated by hypoxia and ADMA. Importantly, the effect of OA-NO2 is accompanied by prevention of STAT3 activation and HIF-1α stabilization. CONCLUSION In summary, OA-NO2 eliminates the manifestation of hypoxia- and ADMA-mediated endothelial dysfunction in HPAEC via the STAT3/HIF-1α cascade. Importantly, our study is bringing a new perspective on molecular mechanisms of NO2-FAs action in pulmonary endothelial dysfunction, which represents a causal link in progression of PH. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Adolf Koudelka
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic
| | - Gabriela Ambrozova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic
| | - Anna Klinke
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.,Department of Experimental Cardiology, University Hospital of Cologne, Kerpener Str. 62, 50924, Cologne, Germany
| | - Tana Fidlerova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic.,Faculty of Science, Institute of Experimental Biology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Hana Martiskova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.,Faculty of Science, Institute of Experimental Biology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Radek Kuchta
- Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3058/10, 616 00, Brno, Czech Republic
| | - Tanja K Rudolph
- Department of Experimental Cardiology, University Hospital of Cologne, Kerpener Str. 62, 50924, Cologne, Germany
| | - Jaroslav Kadlec
- Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3058/10, 616 00, Brno, Czech Republic
| | - Zdenka Kuchtova
- Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3058/10, 616 00, Brno, Czech Republic
| | - Steven R Woodcock
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
| | - Lukas Kubala
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic
| | - Michaela Pekarova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic. .,International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.
| |
Collapse
|
84
|
Kato GJ, Steinberg MH, Gladwin MT. Intravascular hemolysis and the pathophysiology of sickle cell disease. J Clin Invest 2017; 127:750-760. [PMID: 28248201 DOI: 10.1172/jci89741] [Citation(s) in RCA: 460] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hemolysis is a fundamental feature of sickle cell anemia that contributes to its pathophysiology and phenotypic variability. Decompartmentalized hemoglobin, arginase 1, asymmetric dimethylarginine, and adenine nucleotides are all products of hemolysis that promote vasomotor dysfunction, proliferative vasculopathy, and a multitude of clinical complications of pulmonary and systemic vasculopathy, including pulmonary hypertension, leg ulcers, priapism, chronic kidney disease, and large-artery ischemic stroke. Nitric oxide (NO) is inactivated by cell-free hemoglobin in a dioxygenation reaction that also oxidizes hemoglobin to methemoglobin, a non-oxygen-binding form of hemoglobin that readily loses heme. Circulating hemoglobin and heme represent erythrocytic danger-associated molecular pattern (eDAMP) molecules, which activate the innate immune system and endothelium to an inflammatory, proadhesive state that promotes sickle vaso-occlusion and acute lung injury in murine models of sickle cell disease. Intravascular hemolysis can impair NO bioavailability and cause oxidative stress, altering redox balance and amplifying physiological processes that govern blood flow, hemostasis, inflammation, and angiogenesis. These pathological responses promote regional vasoconstriction and subsequent blood vessel remodeling. Thus, intravascular hemolysis represents an intrinsic mechanism for human vascular disease that manifests clinical complications in sickle cell disease and other chronic hereditary or acquired hemolytic anemias.
Collapse
|
85
|
Cannas G, Poutrel S, Thomas X. Hydroxycarbamine: from an Old Drug Used in Malignant Hemopathies to a Current Standard in Sickle Cell Disease. Mediterr J Hematol Infect Dis 2017; 9:e2017015. [PMID: 28293403 PMCID: PMC5333733 DOI: 10.4084/mjhid.2017.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/20/2017] [Indexed: 01/31/2023] Open
Abstract
While hydroxycarbamide (hydroxyurea, HU) has less and fewer indications in malignant hemopathies, it represents the only widely used drug which modifies sickle cell disease pathogenesis. Clinical experience with HU for patients with sickle cell disease has been accumulated over the past 25 years in Western countries. The review of the literature provides increasing support for safety and efficacy in both children and adults for reducing acute vaso-occlusive events including pain episodes and acute chest syndrome. No increased incidence of leukemia and teratogenicity was demonstrated. HU has become the standard-of-care for sickle cell anemia but remains underused. Barriers to its use should be identified and overcome.
Collapse
Affiliation(s)
- Giovanna Cannas
- Hospices Civils de Lyon, Department of Internal Medicine, Edouard Herriot Hospital, Lyon, France
- Claude Bernard University Lyon 1, Laboratoire Interuniversitaire de Biologie de la Motricité EA7424, Equipe ‘Vascular biology and red blood cell’, Villeurbanne, France
| | - Solène Poutrel
- Hospices Civils de Lyon, Department of Internal Medicine, Edouard Herriot Hospital, Lyon, France
| | - Xavier Thomas
- Hospices Civils de Lyon, Hematology Department, Lyon-Sud Hospital, Pierre Bénite, France
| |
Collapse
|
86
|
|
87
|
Weir NA, Saiyed R, Alam S, Conrey A, Desai HD, George MP, Keeley JH, Klings ES, Mehari A, Taylor JG, Minniti CP, Kato GJ. Prostacyclin-analog therapy in sickle cell pulmonary hypertension. Haematologica 2017; 102:e163-e165. [PMID: 28126964 DOI: 10.3324/haematol.2015.131227] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Nargues A Weir
- Cardiovascular and Pulmonary Branch, National Institutes of Health, Bethesda, MD
| | - Rehan Saiyed
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Shoaib Alam
- Cardiovascular and Pulmonary Branch, National Institutes of Health, Bethesda, MD
| | - Anna Conrey
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Himanshu D Desai
- Pulmonary Medicine, Eastern Virginia Medical School, Norfolk, VA
| | - M Patricia George
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, PA
| | - Jennifer H Keeley
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, PA
| | | | - Alem Mehari
- Division of Pulmonary Medicine, Department of Medicine, Howard University, Washington DC, MD
| | - James G Taylor
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Caterina P Minniti
- Division of Hematology, Albert Einstein College of Medicine, Bronx, New York, NY
| | - Gregory J Kato
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, PA, USA
| |
Collapse
|
88
|
Thein MS, Igbineweka NE, Thein SL. Sickle cell disease in the older adult. Pathology 2017; 49:1-9. [PMID: 27914684 PMCID: PMC10757825 DOI: 10.1016/j.pathol.2016.10.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/12/2016] [Accepted: 10/20/2016] [Indexed: 01/14/2023]
Abstract
Sickle cell disease (SCD) is an inherited haemoglobin disorder, associated with recurrent painful episodes, ongoing haemolytic anaemia and progressive multi-organ damage. Until the early 1990s, survival beyond the fourth decade for a patient with SCD was considered unusual and prompted case reports. Nowadays, in countries with developed health care systems, more than 90 percent of newborns with SCD survive into adulthood. Nevertheless, their life expectancy is still shortened by more than two decades compared to the general population. With an increasing life expectancy, SCD has now evolved into a debilitating disorder with substantial morbidity resulting from ongoing sickle cell vasculopathy and multi-organ damage. Limited data on health care issues of older adults with SCD poses multiple challenges to patients, their families and health care providers. In this review, we will address and discuss acute and chronic complications of SCD with a special focus on the older adult.
Collapse
Affiliation(s)
- Mya S Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institute of Health, Bethesda, MD, USA
| | - Norris E Igbineweka
- Department of Haematology, Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institute of Health, Bethesda, MD, USA.
| |
Collapse
|
89
|
Bernaudin F, Pondarré C, Galambrun C, Thuret I. Allogeneic/Matched Related Transplantation for β-Thalassemia and Sickle Cell Anemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1013:89-122. [DOI: 10.1007/978-1-4939-7299-9_4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
90
|
Mathew R, Huang J, Wu JM, Fallon JT, Gewitz MH. Hematological disorders and pulmonary hypertension. World J Cardiol 2016; 8:703-718. [PMID: 28070238 PMCID: PMC5183970 DOI: 10.4330/wjc.v8.i12.703] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/07/2016] [Accepted: 10/09/2016] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH), a serious disorder with a high morbidity and mortality rate, is known to occur in a number of unrelated systemic diseases. Several hematological disorders such as sickle cell disease, thalassemia and myeloproliferative diseases develop PH which worsens the prognosis. Associated oxidant injury and vascular inflammation cause endothelial damage and dysfunction. Pulmonary vascular endothelial damage/dysfunction is an early event in PH resulting in the loss of vascular reactivity, activation of proliferative and antiapoptotic pathways leading to vascular remodeling, elevated pulmonary artery pressure, right ventricular hypertrophy and premature death. Hemolysis observed in hematological disorders leads to free hemoglobin which rapidly scavenges nitric oxide (NO), limiting its bioavailability, and leading to endothelial dysfunction. In addition, hemolysis releases arginase into the circulation which converts L-arginine to ornithine, thus bypassing NO production. Furthermore, treatments for hematological disorders such as immunosuppressive therapy, splenectomy, bone marrow transplantation, and radiation have been shown to contribute to the development of PH. Recent studies have shown deregulated iron homeostasis in patients with cardiopulmonary diseases including pulmonary arterial hypertension (PAH). Several studies have reported low iron levels in patients with idiopathic PAH, and iron deficiency is an important risk factor. This article reviews PH associated with hematological disorders and its mechanism; and iron homeostasis and its relevance to PH.
Collapse
|
91
|
Quinn CT, Smith EP, Arbabi S, Khera PK, Lindsell CJ, Niss O, Joiner CH, Franco RS, Cohen RM. Biochemical surrogate markers of hemolysis do not correlate with directly measured erythrocyte survival in sickle cell anemia. Am J Hematol 2016; 91:1195-1201. [PMID: 27648808 DOI: 10.1002/ajh.24562] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 09/17/2016] [Indexed: 01/18/2023]
Abstract
Hemolysis is a key feature of sickle cell anemia (HbSS). Direct quantitation of hemolysis could be used as an objective outcome in clinical trials of new therapeutics for HbSS and would also enable better human studies of the pathogenesis of complications of HbSS that are ostensibly hemolysis-related, such as pulmonary hypertension. However, contemporary human studies in HbSS have used only surrogate markers of hemolysis rather than direct measurements of RBC survival. We directly quantified hemolysis in HbSS by measuring survival of an age cohort of RBCs labeled with a stable isotope, administered orally as 15 N-glycine, a metabolic precursor of heme. The atomic excess of 15 N in heme extracted from blood was monitored by mass spectrometry over time. We performed 13 labeling experiments in 11 individuals with HbSS. Mean RBC survival was 31.9 days (range 14.1-53.6). Both HbF level, a known determinant of hemolysis, and absolute reticulocyte count (ARC), an index of the marrow's response to hemolysis, correlated with directly measured RBC survival (r = 0.61, P < 0.002; r = -0.84, P < 0.001). However, commonly used biochemical surrogates of hemolysis (LDH, AST, bilirubin, and plasma free hemoglobin) did not correlate with directly measured RBC survival. These biochemical surrogates should be interpreted cautiously, at best, in clinical trials and human physiologic studies in HbSS. ARC was the best correlate of total hemolysis, but only 70% of the variation in RBC survival was reflected in this marker. If greater accuracy is required in human studies, 15 N-glycine RBC labeling can directly and accurately quantify hemolysis. Am. J. Hematol. 91:1195-1201, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Charles T. Quinn
- Division of HematologyCincinnati Children's Hospital Medical CenterCincinnati OH
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnati OH
| | - Eric P. Smith
- Department of Internal MedicineUniversity of Cincinnati College of MedicineCincinnati OH
| | - Shahriar Arbabi
- Department of Internal MedicineUniversity of Cincinnati College of MedicineCincinnati OH
| | - Paramjit K. Khera
- Department of Internal MedicineUniversity of Cincinnati College of MedicineCincinnati OH
| | | | - Omar Niss
- Division of HematologyCincinnati Children's Hospital Medical CenterCincinnati OH
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnati OH
| | - Clinton H. Joiner
- Aflac Cancer and Blood Disorders Center, Emory University School of MedicineAtlanta GA
| | - Robert S. Franco
- Department of Internal MedicineUniversity of Cincinnati College of MedicineCincinnati OH
| | - Robert M. Cohen
- Department of Internal MedicineUniversity of Cincinnati College of MedicineCincinnati OH
- Cincinnati Veterans Affairs Medical CenterCincinnati OH
| |
Collapse
|
92
|
Maron BA, Machado RF, Shimoda L. Pulmonary vascular and ventricular dysfunction in the susceptible patient (2015 Grover Conference series). Pulm Circ 2016; 6:426-438. [PMID: 28090285 PMCID: PMC5210067 DOI: 10.1086/688315] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/21/2016] [Indexed: 12/14/2022] Open
Abstract
Pulmonary blood vessel structure and tone are maintained by a complex interplay between endogenous vasoactive factors and oxygen-sensing intermediaries. Under physiological conditions, these signaling networks function as an adaptive interface between the pulmonary circulation and environmental or acquired perturbations to preserve oxygenation and maintain systemic delivery of oxygen-rich hemoglobin. Chronic exposure to hypoxia, however, triggers a range of pathogenetic mechanisms that include hypoxia-inducible factor 1α (HIF-1α)-dependent upregulation of the vasoconstrictor peptide endothelin 1 in pulmonary endothelial cells. In pulmonary arterial smooth muscle cells, chronic hypoxia induces HIF-1α-mediated upregulation of canonical transient receptor potential proteins, as well as increased Rho kinase-Ca2+ signaling and pulmonary arteriole synthesis of the profibrotic hormone aldosterone. Collectively, these mechanisms contribute to a contractile or hypertrophic pulmonary vascular phenotype. Genetically inherited disorders in hemoglobin structure are also an important etiology of abnormal pulmonary vasoreactivity. In sickle cell anemia, for example, consumption of the vasodilator and antimitogenic molecule nitric oxide by cell-free hemoglobin is an important mechanism underpinning pulmonary hypertension. Contemporary genomic and transcriptomic analytic methods have also allowed for the discovery of novel risk factors relevant to sickle cell disease, including GALNT13 gene variants. In this report, we review cutting-edge observations characterizing these and other pathobiological mechanisms that contribute to pulmonary vascular and right ventricular vulnerability.
Collapse
Affiliation(s)
- Bradley A. Maron
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA; and Department of Cardiology, Boston Veterans Affairs Healthcare System, Boston, Massachusetts, USA
| | - Roberto F. Machado
- Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Larissa Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
93
|
Update on pulmonary arterial hypertension. Rev Clin Esp 2016. [DOI: 10.1016/j.rceng.2016.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
94
|
Schreier DA, Forouzan O, Hacker TA, Sheehan J, Chesler N. Increased Red Blood Cell Stiffness Increases Pulmonary Vascular Resistance and Pulmonary Arterial Pressure. J Biomech Eng 2016; 138:021012. [PMID: 26638883 DOI: 10.1115/1.4032187] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Indexed: 12/13/2022]
Abstract
Patients with sickle cell anemia (SCD) and pulmonary hypertension (PH) have a significantly increased risk of sudden death compared to patients with SCD alone. Sickled red blood cells (RBCs) are stiffer, more dense, more frequently undergo hemolysis, and have a sixfold shorter lifespan compared to normal RBCs. Here, we sought to investigate the impact of increased RBC stiffness, independent of other SCD-related biological and mechanical RBC abnormalities, on the hemodynamic changes that ultimately cause PH and increase mortality in SCD. To do so, pulmonary vascular impedance (PVZ) measures were recorded in control C57BL6 mice before and after ∼50 μl of blood (Hct = 45%) was extracted and replaced with an equal volume of blood containing either untreated RBCs or RBCs chemically stiffened with glutaraldehyde (Hct = 45%). Chemically stiffened RBCs increased mean pulmonary artery pressure (mPAP) (13.5 ± 0.6 mmHg at baseline to 23.2 ± 0.7 mmHg after the third injection), pulmonary vascular resistance (PVR) (1.23 ± 0.11 mmHg*min/ml at baseline to 2.24 ± 0.14 mmHg*min/ml after the third injection), and wave reflections (0.31 ± 0.02 at baseline to 0.43 ± 0.03 after the third injection). Chemically stiffened RBCs also decreased cardiac output, but did not change hematocrit, blood viscosity, pulmonary arterial compliance, or heart rate. The main finding of this study is that increased RBC stiffness alone affects pulmonary pulsatile hemodynamics, which suggests that RBC stiffness plays an important role in the development of PH in patients with SCD.
Collapse
|
95
|
Mehari A, Thomas AV, Thomas AN, Johnson MS. Review: Hemodynamic Characteristics and Outcomes of Sickle Cell Disease Associated Pulmonary Hypertension. Ethn Dis 2016; 26:545-552. [PMID: 27773982 DOI: 10.18865/ed.26.4.545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Pulmonary hypertension (PH) is a leading cause of morbidity and early mortality in adults with sickle cell disease (SCD). However, the prevalence, hemodynamic profile and prognosis of SCD-PH remain controversial and need frequent updates. Pulmonary hypertension determined by right heart catheterization (RHC) occurs in 6% to 10% of adults with SCD. Hemodynamically, SCD-PH may be pre-capillary or post-capillary in nature. The exact etiology is unknown and often multifactorial; hence a thorough diagnostic evaluation following established PH guidelines is essential to determine disease prevalence, etiology and outcomes. Data on the efficacy and safety of pulmonary arterial hypertension (PAH) therapy are limited in SCD; clinical trials in these patients are urgently needed. This review provides an overview of RHC-determined hemodynamic characteristics, current management modality and outcomes; we also highlight recent advances and unmet research needs in SCD-PH.
Collapse
Affiliation(s)
- Alem Mehari
- Division of Pulmonary and Critical Care, Howard University College of Medicine, Washington, DC
| | - Alvin V Thomas
- Division of Pulmonary and Critical Care, Howard University College of Medicine, Washington, DC
| | - Alicia N Thomas
- Division of Pulmonary and Critical Care, Howard University College of Medicine, Washington, DC
| | - Mark S Johnson
- Department of Community and Family Medicine, Howard University College of Medicine, Washington, DC
| |
Collapse
|
96
|
Sarode R, Ballas SK, Garcia A, Kim HC, King K, Sachais B, Williams LA. Red blood cell exchange: 2015 American Society for Apheresis consensus conference on the management of patients with sickle cell disease. J Clin Apher 2016; 32:342-367. [DOI: 10.1002/jca.21511] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/01/2016] [Accepted: 09/13/2016] [Indexed: 01/20/2023]
Affiliation(s)
- Ravi Sarode
- Department of Pathology, Division of Transfusion Medicine and Hemostasis; UT Southwestern Medical Center; Dallas Texas
| | - Samir K. Ballas
- Department of Medicine, Cardeza Foundation for Hematologic Research; Jefferson Medical College, Thomas Jefferson University; Philadelphia Pennsylvania
| | - Alicia Garcia
- Children's Hospital & Research Center; Oakland California
| | - Haewon C Kim
- Departments of Pediatrics and Pathology; Children's Hospital of Philadelphia, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA
| | - Karen King
- Department of Pathology, Transfusion Medicine; Johns Hopkins University; Baltimore Maryland
| | | | - Lance A. Williams
- Department of Pathology, Division of Laboratory Medicine; University of Alabama at Birmingham; Birmingham Alabama
| |
Collapse
|
97
|
Geard A, Pule GD, Chelo D, Bitoungui VJN, Wonkam A. Genetics of Sickle Cell-Associated Cardiovascular Disease: An Expert Review with Lessons Learned in Africa. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 20:581-592. [PMID: 27726639 PMCID: PMC5067873 DOI: 10.1089/omi.2016.0125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sickle cell disease (SCD) vastly impacts the African continent and is associated with cardiovascular diseases. Stroke, kidney disease, and pulmonary hypertension are considered as proxies of severity in SCD with several genomic loci implicated in their heritability. The present expert review examined the current data on epidemiology and genetic risk factors of stroke, pulmonary hypertension, and kidney disease associated with SCD, as indexed in PubMed® and Google Scholar®. Studies collectively show that stroke and kidney disease each affect ∼10% of SCD patients, with pulmonary hypertension displaying a higher prevalence of 30% among adults with SCD. There is some evidence that these epidemiology figures may be an underestimate in SCD patients living in Africa. A modest number of publications have identified genetic factors involved in pathways regulating inflammation, coagulation, cell adhesion, heme degradation, α-globin and γ-globin production, and others, which contribute to the development risk of targeted cardiovascular phenotypes. However, in most cases, these studies have not been validated across populations. There is therefore an urgent need for large-scale genome-wide association, whole-exome and whole-genome studies, and multiomics research on cardiovascular diseases associated with SCD, particularly in Africa, to allow for proportional investment of global research funding on diseases that greatly impact the African continent. Ultimately, this will cultivate socially responsible research investments and identification of at-risk individuals with improved preventive medicine, which should be a cornerstone of global precision medicine.
Collapse
Affiliation(s)
- Amy Geard
- Division of Human Genetics, Departments of Medicine and Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gift D. Pule
- Division of Human Genetics, Departments of Medicine and Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - David Chelo
- Department of Paediatrics, Faculty of Medicine and Biomedical Sciences, University of Yaoundé, Yaoundé, Cameroon
| | | | - Ambroise Wonkam
- Division of Human Genetics, Departments of Medicine and Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
98
|
Doppler-Defined Pulmonary Hypertension in Sickle Cell Anemia in Kurdistan, Iraq. PLoS One 2016; 11:e0162036. [PMID: 27583566 PMCID: PMC5008799 DOI: 10.1371/journal.pone.0162036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 08/16/2016] [Indexed: 02/02/2023] Open
Abstract
To determine the frequency, clinical and laboratory associations of pulmonary hypertension in Iraqi Kurds with sickle cell anemia, a total of ninety four such patients attending a major hemoglobinopathy center in Iraqi Kurdistan were enrolled. All patients were re-evaluated clinically and had their blood counts, HbF, serum ferritin, LDH, renal and liver function assessed. Transthoracic Doppler echocardiography with measurement of tricuspid valve regurgitant jet velocity (TRV) was performed. A TRV in excess of 2.8 m/s was considered for the purposes of this study as indicative of pulmonary hypertension (PH). The prevalence of TRV in excess of 2.8m/s was 10.6%. By univariate analysis: significantly higher reticulocyte count, more frequent blood transfusions and pain episodes were encountered in the PH group as compared to the non-PH group (p = 0.001, 0.045 and 0.02 respectively). Moreover, PH patients had significantly higher mean right atrial area, left atrial size, E wave/A wave ratio and ejection fraction by echocardiography (p = 0.027, 0.037, <0.001 and 0.008 respectively). Except for reticulocyte count none of the other parameters remained significant by multivariate analysis (p = 0.024). In conclusion the current study revealed that pulmonary hypertension is rather frequent among Iraqi Kurds with sickle cell anemia, and identified reticulocyte count as an independently associated parameter with PH in this population. Future prospective studies including right heart catheterization and appropriate medical intervention are warranted.
Collapse
|
99
|
Bakeer N, James J, Roy S, Wansapura J, Shanmukhappa SK, Lorenz JN, Osinska H, Backer K, Huby AC, Shrestha A, Niss O, Fleck R, Quinn CT, Taylor MD, Purevjav E, Aronow BJ, Towbin JA, Malik P. Sickle cell anemia mice develop a unique cardiomyopathy with restrictive physiology. Proc Natl Acad Sci U S A 2016; 113:E5182-E5191. [PMID: 27503873 PMCID: PMC5024607 DOI: 10.1073/pnas.1600311113] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cardiopulmonary complications are the leading cause of mortality in sickle cell anemia (SCA). Elevated tricuspid regurgitant jet velocity, pulmonary hypertension, diastolic, and autonomic dysfunction have all been described, but a unifying pathophysiology and mechanism explaining the poor prognosis and propensity to sudden death has been elusive. Herein, SCA mice underwent a longitudinal comprehensive cardiac analysis, combining state-of-the-art cardiac imaging with electrocardiography, histopathology, and molecular analysis to determine the basis of cardiac dysfunction. We show that in SCA mice, anemia-induced hyperdynamic physiology was gradually superimposed with restrictive physiology, characterized by progressive left atrial enlargement and diastolic dysfunction with preserved systolic function. This phenomenon was absent in WT mice with experimentally induced chronic anemia of similar degree and duration. Restrictive physiology was associated with microscopic cardiomyocyte loss and secondary fibrosis detectable as increased extracellular volume by cardiac-MRI. Ultrastructural mitochondrial changes were consistent with severe chronic hypoxia/ischemia and sarcomere diastolic-length was shortened. Transcriptome analysis revealed up-regulation of genes involving angiogenesis, extracellular-matrix, circadian-rhythm, oxidative stress, and hypoxia, whereas ion-channel transport and cardiac conduction were down-regulated. Indeed, progressive corrected QT prolongation, arrhythmias, and ischemic changes were noted in SCA mice before sudden death. Sudden cardiac death is common in humans with restrictive cardiomyopathies and long QT syndromes. Our findings may thus provide a unifying cardiac pathophysiology that explains the reported cardiac abnormalities and sudden death seen in humans with SCA.
Collapse
MESH Headings
- Anemia, Sickle Cell/complications
- Anemia, Sickle Cell/physiopathology
- Animals
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/physiopathology
- Cardiomyopathies/etiology
- Cardiomyopathies/genetics
- Cardiomyopathies/physiopathology
- Death, Sudden, Cardiac/etiology
- Disease Models, Animal
- Electrocardiography/methods
- Gene Expression Profiling
- Heart Failure, Diastolic/etiology
- Heart Failure, Diastolic/genetics
- Heart Failure, Diastolic/physiopathology
- Humans
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/physiopathology
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Myocardium/metabolism
- Myocardium/pathology
Collapse
Affiliation(s)
- Nihal Bakeer
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Jeanne James
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Swarnava Roy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Janaka Wansapura
- Imaging Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Department of Physics, University of Colombo, Colombo 03, Sri Lanka
| | | | - John N Lorenz
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH 45267
| | - Hanna Osinska
- Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Kurt Backer
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Anne-Cecile Huby
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Archana Shrestha
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Omar Niss
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Robert Fleck
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Charles T Quinn
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Michael D Taylor
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Enkhsaikhan Purevjav
- Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Bruce J Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Jeffrey A Towbin
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Punam Malik
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229;
| |
Collapse
|
100
|
Soh MC, Sankaran S, Chung NY, Nelson-Piercy C, Howard J, Robinson SE, Oteng-Ntim E. Mildly raised tricuspid regurgitant velocity 2.5-3.0 m/s in pregnant women with sickle cell disease is not associated with poor obstetric outcome - An observational cross-sectional study. Obstet Med 2016; 9:160-163. [PMID: 27829875 DOI: 10.1177/1753495x16651529] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/04/2016] [Indexed: 02/02/2023] Open
Abstract
Pulmonary hypertension is associated with 36% mortality in pregnancy, and 6-10% of patients with sickle cell disease have pulmonary hypertension. Tricuspid regurgitant velocity ≥2.5 m/s on echocardiography is a well validated means of screening for pulmonary hypertension in the non-pregnant population. This is a pilot study to determine if this is a useful non-invasive screening test for pulmonary hypertension in pregnancy, and whether raised tricuspid regurgitant velocity ≥2.5 m/s was associated with poor outcomes. This is a cross-sectional study over a five-year period in a tertiary referral centre with a specialised multidisciplinary clinic for pregnant women with sickle cell disease. Women with sickle cell disease, no prior pulmonary hypertension and singleton pregnancies who had echocardiography with a measurable tricuspid regurgitant velocity in pregnancy were included. There were 34 pregnancies, of which eight had tricuspid regurgitant velocity ≥2.5 m/s. There were no significant differences in their characteristics, sickle cell-related complications or medical co-morbidities. The women with tricuspid regurgitant velocity ≥2.5 m/s had similar obstetric and perinatal outcomes as those with a tricuspid regurgitant velocity <2.5 m/s.
Collapse
Affiliation(s)
- May C Soh
- Women's Health Academic Department, King's College London, Guy's and St Thomas' Hospital NHS Foundation Trust, London, UK
| | - Srividhya Sankaran
- Women's Health Academic Department, King's College London, Guy's and St Thomas' Hospital NHS Foundation Trust, London, UK
| | - Natali Ya Chung
- Cardiology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Catherine Nelson-Piercy
- Women's Health Academic Department, King's College London, Guy's and St Thomas' Hospital NHS Foundation Trust, London, UK
| | - Jo Howard
- Haematology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Sue E Robinson
- Haematology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Eugene Oteng-Ntim
- Women's Health Academic Department, King's College London, Guy's and St Thomas' Hospital NHS Foundation Trust, London, UK
| |
Collapse
|